A variational framework for accelerated optimization was recently introduced on normed vector spaces and Riemannian manifolds in [
Citation: Valentin Duruisseaux, Melvin Leok. Time-adaptive Lagrangian variational integrators for accelerated optimization[J]. Journal of Geometric Mechanics, 2023, 15(1): 224-255. doi: 10.3934/jgm.2023010
[1] | Silpak Biswas, Rintu Das, Ena Ray Banerjee . Role of free radicals in human inflammatory diseases. AIMS Biophysics, 2017, 4(4): 596-614. doi: 10.3934/biophy.2017.4.596 |
[2] | Sonia Shastri, Ravichandra Vemuri, Nuri Gueven, Madhur D. Shastri, Rajaraman Eri . Molecular mechanisms of intestinal inflammation leading to colorectal cancer. AIMS Biophysics, 2017, 4(1): 152-177. doi: 10.3934/biophy.2017.1.152 |
[3] | Armenuhi Moghrovyan, Naira Sahakyan . Antimicrobial activity and mechanisms of action of Origanum vulgare L. essential oil: effects on membrane-associated properties. AIMS Biophysics, 2024, 11(4): 508-526. doi: 10.3934/biophy.2024027 |
[4] | Sweta Raikundalia, Ling Ling Few, Siti Asma' Hassan, Get Bee Yvonne-Τee, Wei Cun See Too . Choline kinase and miR-32-5p: A crucial interaction promoting apoptosis and delaying wound repair in cervical cancer cells. AIMS Biophysics, 2024, 11(3): 281-295. doi: 10.3934/biophy.2024016 |
[5] | Francesca Ballarini, Mario P. Carante, Alessia Embriaco, Ricardo L. Ramos . Effects of ionizing radiation in biomolecules, cells and tissue/organs: basic mechanisms and applications for cancer therapy, medical imaging and radiation protection. AIMS Biophysics, 2022, 9(2): 108-112. doi: 10.3934/biophy.2022010 |
[6] | G.Corradini Maria, LaviniaWang Yan, Le An, M.Waxman Sarah, Zelent Bogumil, Chib Rahul, Gryczynski Ignacy, D.Ludescher Richard . Identifying and selecting edible luminescent probes as sensors of food quality. AIMS Biophysics, 2016, 3(2): 319-339. doi: 10.3934/biophy.2016.2.319 |
[7] | Min Zhang, Shijun Lin, Wendi Xiao, Danhua Chen, Dongxia Yang, Youming Zhang . Applications of single-cell sequencing for human lung cancer: the progress and the future perspective. AIMS Biophysics, 2017, 4(2): 210-221. doi: 10.3934/biophy.2017.2.210 |
[8] | Manuel Bedrossian, Marwan El-Kholy, Daniel Neamati, Jay Nadeau . A machine learning algorithm for identifying and tracking bacteria in three dimensions using Digital Holographic Microscopy. AIMS Biophysics, 2018, 5(1): 36-49. doi: 10.3934/biophy.2018.1.36 |
[9] | Reza Hadjiaghaie Vafaie, Aysan Madanpasandi . In-situ AC electroosmotic and thermal perturbation effects for wide range of ionic strength. AIMS Biophysics, 2017, 4(3): 451-464. doi: 10.3934/biophy.2017.3.451 |
[10] | Larisa I. Fedoreyeva, Boris F. Vanyushin . Regulation of DNA methyltransferase gene expression by short peptides in nicotiana tabacum regenerants. AIMS Biophysics, 2021, 8(1): 66-79. doi: 10.3934/biophy.2021005 |
A variational framework for accelerated optimization was recently introduced on normed vector spaces and Riemannian manifolds in [
Photobiomodulation (PBM) was discovered almost 50 years ago by Endre Mester in Hungary [1]. For most of this time PBM was known as "low-level laser therapy" as ruby laser (694 nm) and HeNe lasers (633 nm) were the first devices used. Recently a consensus decision [2] was taken to use the terminology "PBM" since the term "low-level" was very subjective, and it is now known that actual lasers are not required, as non-coherent light-emitting diodes (LEDs) work equally well [3]. For much of this time the mechanism of action of PBM was unclear, but in recent years much progress has been made in elucidating chromophores and signaling pathways [4].
Most of the early work in this field was carried out with various kinds of lasers, and it was thought that laser light had some special characteristics not possessed by light from other light sources such as sunlight, fluorescent or incandescent lamps and now LEDs. However all the studies that have been done comparing lasers to equivalent light sources with similar wavelength and power density of their emission, have found essentially no difference between them.
Many wavelengths in the red (600–700 nm) and near-infrared (NIR, 770–1200 nm) spectral regions have shown positive results, however there is a region in between (700–770 nm) where broadly speaking, the results are likely to be disappointing. Recently blue and green wavelengths have also begun to be explored [5] but they have major problems with penetration depth. It is accepted that penetration of light into tissue is governed by both absorption and scattering by molecules and structures present in tissue. Both absorption and scattering become significantly less as the wavelength gets longer, so the penetration depth of NIR is maximal about 810 nm, and at longer wavelengths water becomes an important absorber and penetration depth gets shorter again [6].
The "biphasic dose response" describes a situation in which there is an optimum value of the "dose" of PBM most often defined by the energy density (J/cm2) [7,8]. It has been consistently found that when the dose of PBM is increased a maximum response is reached at some value, and if the dose in increased beyond that maximal value, the response diminishes, disappears and it is even possible that negative or inhibitory effects are produced at very high fluences.
Cytochrome c oxidase (CCO) is unit Ⅳ in the mitochondrial electron transport chain. It transfers one electron (from each of four cytochrome c molecules), to a single oxygen molecule, producing two molecules of water. At the same time the four protons required, are translocated across the mitochondrial membrane, producing a proton gradient that the ATP synthase enzyme needs to synthesize ATP. CCO has two heme centers (a and a3) and two copper centers (CuA and CuB). Each of these metal centers can exist in an oxidized or a reduced state, and these have different absorption spectra, meaning CCO can absorb light well into the NIR region (up to 950 nm) [9]. Tiina Karu from Russia was the first to suggest [10,11], that the action spectrum of PBM effects matched the absorption spectrum of CCO, and this observation was confirmed by Wong-Riley et al in Wisconsin [12]. The assumption that CCO is a main target of PBM also explains the wide use of red/NIR wavelengths as these longer wavelengths have much better tissue penetration than say blue or green light which are better absorbed by hemoglobin. The most popular theory to explain exactly why photon absorption by CCO could led to increase of the enzyme activity, increased oxygen consumption, and increased ATP production is based on photodissociation of inhibitory nitric oxide (NO) [13]. Since NO is non-covalently bound to the heme and Cu centers and competitively blocks oxygen at a ratio of 1:10, a relatively low energy photon can kick out the NO and allow a lot of respiration to take place [14].
More recently it has become apparent that another class of photoreceptors, must be involved in transducing cellular signals, particularly responding to blue and green light. Thee photoreceptors have been proposed to be members of the family of light-sensitive G-protein coupled receptors known as opsins (OPN). Opsins function by photoisomerization of a cis-retinal co-factor leading to a conformational change in the protein. The most well known opsin is rhodopsin (OPN1), which is responsible for mediating vision in the rod and cone photoreceptor cells in the mammalian retina. There are other members of the opsin family (OPN2-5), which are expressed in many other tissues of the body including the brain [15]. One of the best-defined signaling events that occurs after light-activation of opsins, is the opening of light-gated ion channels such as members of the transient receptor potential (TRP) family of calcium channels [16]. TRP channels are now known to be pleiotropic cellular sensors mediating the response to a wide range of external stimuli (heat, cold, pressure, taste, smell), and involved in many different cellular processes [17]. Activation of TRP causes non-selective permeabilization (mainly of the plasma membrane) to calcium, sodium and magnesium [18]. It is now known that TRP channel proteins are conserved throughout evolution and are found in most organisms, tissues, and cell-types. The TRP channel superfamily is now classified into seven related subfamilies: TRPC, TRPM, TRPV, TRPA, TRPP, TRPML, and TRPN [19]. Light-sensitive ion channels are based on an opsin chromophore (isomerization of a cis-retinal molecule to the trans configuration) as illustrated in Drusophila photoreceptors [20].
We have shown that blue or green light (but not red or 810 nm NIR) increased intracellular calcium in adipose derived stem cells, that could be blocked by ion channel inhibitors [5].
There is another well-known family of biological chromophores called cryptochromes. These proteins have some sequence similarity to photolyases [21], which are blue light responsive enzymes that repair DNA damage in bacteria caused by UV exposure [22]. Cryptochromes rely on a flavin (flavin adenine dinucleotide, FAD) or a pterin (5, 10-methenyltetrahydrofolic acid) to actually absorb the light (again usually blue or green). Cryptochromes have been studied mainly in plants and insects. Recent evidence has emerged that mammalian cryptochromes are important in regulation of the circadian clock. It is thought that human cryptochromes (CRY1 and CRY2) send signals via part of the optic nerve to the suprachiasmatic nucleus (SCN) in the brain, which is the master regulator of the CLOCK system to entrain biological responses to the light-dark cycle [23]. However the situation is complicated because retinal ganglion cells containing melanopsin (OPN4) are also involved in photoentrainment [24]. Studies are still ongoing to investigate this redundancy [25].
It should be emphasized that compared to CCO and mitochondria, evidence is still emerging concerning the extent to which opsins, cryptochomes and light-gated ion channels (which may be widely expressed in many different cell types) could be responsible for PBM effects. If their role is significant it is likely to be in the blue and green spectral regions. Further research will be necessary to explore their role in anti-inflammatory effects, wound healing and tissue regeneration.
Since the biological effects of light continue to be observed, as the wavelength increases in the infra-red region (>1000 nm), beyond those known to be absorbed by CCO, it is now thought likely that an alternative chromophore must be responsible. The obvious candidate for this alternative chromophore is water molecules whose absorption spectrum has peaks at 980 nm, and also at most wavelengths longer than 1200 nm. Moreover, water is by the far the most prevalent molecule in biological tissue (particularly considering its low molecule weight = 18). At present the proposed mechanism involves selective absorption of IR photons by structured water layers (also known as interfacial water) [26] or water clusters [27], at power levels that are insufficient to cause any detectable bulk-heating of the tissue. A small increase in vibrational energy by a water cluster formed in or on a sensitive protein such as a heat-gated ion channel, could be sufficient to perturb the tertiary protein structure thus opening the channel and allowing modulation of intracellular calcium levels [28]. Pollack has shown that interfacial water can undergo charge separation when it absorbs visible or NIR light [29]. This charge separation (equivalent to localized pH changes) could affect the conformation of proteins [30]. It has also been suggested that PBM could reduce the viscosity of interfacial water within the mitochondria, and allow the F0F1 ATP synthase, which rotates as a nanomotor to turn faster [31]. It should be noted here that the first regulatory approvals of PBM were gained as a 510 K device "equivalent to an non-heating IR lamp" [32]. While the involvement of water as a chromophore may still be considered hypothetical it is difficult to think of another explanation for the beneficial of PBM at wavelengths between 1000 nm all the way to 10, 000 nm (carbon dioxide laser).
The molecular chromophores discussed above are graphically summarized in Figure 1.
When PBM stimulates CCO activity in normal healthy cells, the resulting increase in mitochondrial membrane potential (MMP) above normal baseline levels, leads to a brief and rather modest increase in generation of reactive oxygen species (ROS) [33]. However this brief burst of ROS caused by 3 J/cm2 of 810 nm laser (Figure 2A) was shown to be sufficient to activate the redox-sensitive transcription factor, NF-kB in embryonic fibroblasts [34] (Figure 2B). Addition of the anti-oxidant N-acetyl-cysteine to the cells could block the NK-kB activation (Figure 2C), but not the increase in cellular ATP caused by the mitochondrial stimulation (Figure 2D). In primary cultured cortical neurons [35], 810 nm laser produced a biphasic dose response in ATP production (Figure 3A) and MMP (Figure 3B) with a maximum at 3 J/cm2. At a high dose (30 J/cm2) the MMP was actually lowered below baseline. Interestingly the dose-response curve between fluence (J/cm2) and ROS production showed two different maxima (Figure 3C). One of these maxima occurred at 3 J/cm2 where the MMP showed its maximum increase. The second maximum in ROS production occurred at 30 J/cm2 where the MMP had been reduced below baseline. At a value between these two fluences (10 J/cm2) a dose at which the MMP was approximately back to baseline, there was not much ROS generation. These data are very good examples of the "biphasic dose response" or "Arndt-Schulz curve" which is often discussed in the PBM literature [7,8].
Thus it appears that ROS can be generated within mitochondria when the MMP is increased above normal values and also when it is decreased below normal values. It remains to be seen whether these two kinds of PBM-generated ROS are identical or not. One intriguing possibility is that whether the ROS generated by PBM is beneficial or detrimental may depend on the rate at which it is generated. If superoxide is generated in mitochondria at a rate that allows superoxide dismutase (SOD) to detoxify it to hydrogen peroxide, then the uncharged H2O2 can diffuse out of the mitochondria to activate beneficial signaling pathways, while if superoxide is generated at a rate or at levels beyond the ability of SOD to deal with it, then the charged superoxide may build up inside mitochondria and damage them.
Notwithstanding, the ability of PBM to produce a burst of ROS in normal cells, it is well-accepted that PBM when as a treatment for tissue injury or muscle damage is able to reduce markers of oxidative stress [36,37,38]. How can these apparently contradictory findings be reconciled? A study attempted to answer this question [39]. Primary cultured cortical neurons were treated with one of three different interventions, all of which were chosen from literature methods of artificially inducing oxidative stress in cell culture. The first was cobalt chloride (CoCl2), which is used as a mimetic for hypoxia and works by a Fenton reaction producing hydroxyl radicals [40]. The second was direct treatment with hydrogen peroxide. The third was treatment with the mitochondrial complex Ⅰ inhibitor, rotenone [41]. All three of these different treatments increased the intracellular mitochondrial ROS as judged by Cell-Rox Red (Figure 4A), and at the same time lowered the MMP as measured by tetramethyl-rhodamine methyl ester (TMRM) (Figure 4B). PBM (3 J/cm2 of 810 nm laser) raised the MMP back towards baseline, while simultaneously reducing the generation of ROS in oxidatively stressed cells (while slightly increasing ROS in normal cells). In control cells (no oxidative stress), PBM increased MMP above baseline and still produced a modest increase in ROS.
Since most laboratory studies of PBM as a therapy have looked at various animal models of disease or injury, it is not surprising that most workers have measured reduction in tissue markers of oxidative stress (TBARS) after PBM [36,42]. There have been a lot of studies looking at muscles. In humans, especially in athletes, high-level exercise produces effects in muscles characterized by delayed-onset muscle soreness, markers of muscle damage (creatine kinase), inflammation and oxidative stress.
One cellular study by Macedo et al [43] used muscle cells isolated from muscular dystrophy mice (mdx LA 24) and found that 5 J/cm2 of 830 nm increased the expression levels of myosin heavy chain, and intracellular [Ca2+]i. PBM decreased H2O2 production and 4-HNE levels and also GSH levels and GR and SOD activities. The mdx cells showed significant increase in the TNF-α and NF-κB levels, which were reduced by PBM.
While it is highly likely that the effects of PBM in modulating ROS are involved in the anti-inflammatory effects of PBM, it would be dangerous to conclude that that is the only explanation. Other signaling pathways (nitric oxide, cyclic AMP, calcium) are also likely to be involved in reduction of inflammation.
As mentioned above we found [34] that PBM (3 J/cm2 of 810 nm laser) activated NF-kB in embryonic fibroblasts isolated from mice that had been genetically engineered to express firefly luciferase under control of an NF-kB promoter. Although it is well-known that NF-kB functions as a pro-inflammatory transcription factor, but on the other hand it is also well known that in clinical practice or in laboratory animal studies) PBM has a profound anti-inflammatory effect in vivo. This gives rise to another apparent contradiction that must be satisfactorily resolved.
Part of the answer to the apparent contradiction highlighted above, was addressed in a subsequent paper [44]. We isolated primary bone marrow-derived dendritic cells (DCs) from the mouse femur and cultured them with GM-CSF. When these cells were activated with the classical toll-like receptor (TLR) agonists, LPS (TLR4) and CpG oligodeoxynucleotide (TLR9), they showed upregulation of cell-surface markers of activation and maturation such as MHC class Ⅱ, CD86 and CD11c as measured by flow cytometry. Moreover IL12 was secreted by CpG-stimulated DCs. PBM (0.3 or 3 J/cm2 of 810 nm laser) reduced all the markers of activation and also the IL12 secretion. Figure 5.
Yamaura et al [45] tested PBM (810 nm, 5 or 25 J/cm2) on synoviocytes isolated from rheumatoid arthritis patients. They applied PBM before or after addition of tumor necrosis factor-α (TNF-α). mRNA and protein levels of TNF-α and interleukins (IL)-1beta, and IL-8 were reduced (especially by 25 J/cm2).
Hwang et al [46] incubated human annulus fibrosus cells with conditioned medium obtained from macrophages (THP-1 cells) containing proinflammatory cytokines IL1β, IL6, IL8 and TNF-α. They compared 405, 532 and 650 nm at doses up to 1.6 J/cm2. They found that all wavelengths reduced IL8 expression and 405 nm also reduced IL6.
The "Super-Lizer" is a Japanese device that emits linear polarized infrared light. Imaoka et al [47] tested it against a rat model of rheumatoid arthritis involving immunizing the rats with bovine type Ⅱ collagen, after which they develop autoimmune inflammation in multiple joints. The found reductions in IL20 expression in histological sections taken from the PBM-treated joints and also in human rheumatoid fibroblast-like synoviocyte (MH7A) stimulated with IL1β.
Lim et al [48] studied human gingival fibroblasts (HGF) treated with lipopolysaccharides (LPS) isolated from Porphyromonas gingivalis. They used PBM mediated by a 635 nm LED and irradiated the cells + LPS directly or indirectly (transferring medium from PBM treated cells to other cells with LPS). Both direct and indirect protocols showed reductions in inflammatory markers (cyclooxygenase-2 (COX2), prostaglandin E2 (PGE2), granulocyte colony-stimulating factor (G-CSF), regulated on activated normal T-cell expressed and secreted (RANTES), and CXCL11). In the indirect irradiation group, phosphorylation of C-Raf and Erk1/2 increased. In another study [49] the same group used a similar system (direct PBM on HGF + LPS) and showed that 635 nm PBM reduced IL6, IL8, p38 phosphorylation, and increased JNK phosphorylation. They explained the activation of JNK by the growth promoting effects of PBM. Sakurai et al reported [50] similar findings using HGF treated with Campylobacter rectus LPS and PBM (830 nm up to 6.3 J/cm2) to reduce levels of COX2 and PGE2. In another study [51] the same group showed a reduction in IL1β in the same system.
Another very interesting property of PBM is its ability to change the phenotype of activated cells of the monocyte or macrophage lineage. These cells can display two very different phenotypes depending on which pathological situation the cells are faced with. The M1 phenotype (classically activated) applies to macrophages that are faced with a situation in which bacteria or other pathogens need to be killed, or alternatively tumor cells need to be destroyed. Inducible nitric oxide synthase is a hallmark of the M1 phenotype and nitric oxide secretion is often measured. On the other hand the M2 phenotype (alternatively activated) applies to macrophages that are involved in disposal of cellular or protein debris and stimulation of healing by angiogenesis. The M2 phenotype produces arginase, an enzyme that inhibits NO production and allows them to produce ornithine, a precursor of hydroxyproline and polyamines [52]. The markers of these two phenotypes of activated macrophage have some aspects in common, but also show many aspects that are very different [53]. It should be noted that this concept of M1 and M2 activation states, applies to other specialized macrophage type cells that are resident in different tissues, such as microglia in brain [54], alveolar macrophages in lung [55], Kuppfer cells in liver [56], etc.
Fernandes et al used J774 macrophage-like cells activated with interferon-γ and LPS to produce a MI phenotype and compared 660 nm and 780 nm laser. They found that both wavelengths reduced TNF-α, COX-2 and iNOS expression, with the 780 nm being somewhat better [57]. Silva et al used RAW264.7 macrophages to test two wavelengths (660 nm and 808 nm) at a range of fluences (11-214 J/cm2) [58]. They found increases in NO release with 660 nm at the higher fluences. von Leden et al carried out an interesting study looking at the effects of PBM on microglia and their interaction with cortical neurons [59]. They used both primary microglia isolated from mouse brains and the BV2 mouse microglial cell line and compared four fluences (0.2, 4, 10, and 30 J/cm2, at 808 nm. Fluences between 4 and 30 J/cm2 induced expression of M1 markers in microglia. Markers of the M2 phenotype, including CD206 and TIMP1, were observed at lower energy densities of 0.2–10 J/cm2. In addition, co-culture of PBM or control-treated microglia with primary neuronal cultures demonstrated a dose-dependent effect of PBM on microglial-induced neuronal growth and neurite extension. This suggests that the benefits of PBM on neuroinflammation may be more pronounced at lower overall doses. The same group went on to show that M1 activated macrophages receiving PBM (660 nm laser) showed significant decreases in CCL3, CXCL2 and TNFα mRNA expression 4 h after irradiation [60]. However, 24 h after irradiation, M1 macrophages showed increased expression of CXCL2 and TNFα genes. M1 activated macrophages irradiated with 780 nm showed a significant decrease in CCL3 gene expression 4h after irradiation. These data could explain the anti-inflammatory effects of LLLT in wound repair.
This section will cover some of the most important medical indications where PBM has been shown in laboratory studies to be effective (at least partly) by its pronounced anti-inflammatory effects. Figure 6 shows a graphical summary of the anti-inflammatory applications of PBM in experimental animal models.
Many papers have demonstrated the efficacy of PBM in stimulating wound healing. In animal models these studies have generally been on acute wounds [61], while in clinical trials they are often been concerned with chronic non-healing wounds such as diabetic ulcers [62]. Gupta et al [63] tested PBM using a superpulsed 904 nm laser on burn wounds in rats. They found faster healing, reduced inflammation (histology), decreased expression of TNF-α and NF-kB, and up-regulated expression of VEGF, FGFR-1, HSP-60, HSP-90, HIF-1α and matrix metalloproteinases-2 and 9 compared to controls. It is intriguing to speculate that the effects of PBM on wound healing (especially the use of for chronic non-healing wounds) could involve both pro-inflammatory effects and anti-inflammatory effects. This seemingly contradictory statement may be possible due to the recent discovery of resolvins and protectins, which are multifunctional lipid mediators derived from omega-3 polyunsaturated fatty acids [64]. If resolvins were produced as a result of the brief acute inflammation induced by application of PBM to chronic wounds, then it has been already shown that resolvins can hasten the healing of diabetic wounds in mice [65]. Resolvins have been shown to reduce tumor necrosis factor-α, interleukin-1β, and neutrophil platelet-endothelial cell adhesion molecule-1 in a mouse burn wound model [66].
In humans, arthritis is most often caused by a degenerative process occurring in osteoarthritis, or an autoimmune process occurring in rheumatoid arthritis. Both are characterized by pronounced inflammatory changes in the joint and even systemically. Different animal models are produced to mimic these diseases, but a common approach is to inject the sterile preparation of yeast cell walls known as zymosan into the knee joints of rats.
Castano et al [67] used this zymosan-induced arthritis model to study the effects of two different fluences of 810 nm laser (3 and 30 J/cm2) delivered at two different power densities (5 and 50 mW/cm2). PBM was delivered once a day for 5 days commencing after zymosan injection, and the swelling in the knee was measured daily. Prostagladin E2 (PGE2) was measured in the serum. They found that 3 out of the 4 sets of parameters were approximately equally effective in reducing swelling and PGE2, but the ineffective set of parameters was 3 J/cm2 delivered at 50 mW/cm2 which only took 1 min of illumination time. The conclusion was, that the illumination time was important in PBM, and if that time was too short, then the treatment could be ineffective.
Moriyama et al [68] used a transgenic mouse strain (FVB/N-Tg (iNOS-luc) that had been engineered to express luciferase under control of the inducible nitric oxide synthase promoter, to allow bioluminescence imaging of PBM of the zymosal-induced arthritis model in mice knees. They compared the same fluence of 635, 660, 690, and 905 nm (CW0 and 905 nm (short pulse). Animals younger than 15 weeks showed mostly reduction of iNOS expression, while older animals showed increased iNOS expression. Pulsed 905 nm also increased iNOS expression.
Pallotta et al [69] used a model where carageenan was injected into the rat knee and tested 810 nm laser at 1, 3, 6 or 10 J/cm2. Rats were sacrificed after 6 or 12 hours and the joint tissue removed. PBM was able to significantly inhibit the total number of leukocytes, as well as the myeloperoxidase activity. Vascular extravasation was significantly inhibited at the higher dose of energy of 10 J. Gene expression of both COX-1 and 2 were significantly enhanced by laser irradiation while PGE2 production was inhibited. These apparently contradictory results require more study to fully explain.
One of the most robust applications of PBM, is its effects on muscles [70,71]. PBM can potentiate muscular performance especially when applied to the muscles 3 hours before exercise [72]. PBM can also make exercise-training regimens more effective. It is not therefore surprising that PBM can also help to heal muscle injuries, not to mention reducing muscle pain and soreness after excessive exercise. Many of the animal studies that have been done have looked at markers of inflammation and oxidative stress in muscle tissue removed from sacrificed animals. For instance, Silveira et al [73] caused a traumatic muscle injury by a single blunt-impact to the rat gastrocnemius muscle. PBM (850 nm, 3 or 5 J/cm2) was initiated 2, 12, and 24 h after muscle trauma, and repeated for five days. The locomotion and muscle function was improved by PBM. TBARS, protein carbonyls, superoxide dismutase, glutathione peroxidase, and catalase, were increased after muscle injury, these increases were prevented by PBM. PBM prevented increases in IL-6 and IL-10 and reversed the trauma-induced reduction in BDNF and VEGF.
There have been many studies that have looked at the effects of PBM on pain in animal models. Some studies have looked at sensitivity to pain [74] using the von Frey filaments (a graded set of fibers of increasing stiffness and when the animal feels the pressure it withdraws its foot [75]).
Some studies have looked at animal models of neuropathic pain such as the "spared nerve injury" [76]. This involves ligating two out of three branches of the sciatic nerve in rats and causes long lasting (>6 months) mechanical allodynia [77]. Kobelia Ketz et al found improvements in pain scores with PBM (980 nm applied to affected hind paw 1 W, 20 s, 41 cm above skin, power density 43.25 mW/cm2, dose 20 J). They also found lower expression of the proinflammatory marker (Iba1) in microglia in the dorsal root ganglion, gracile nucleus, dorsal column and dorsal horn. The M1/M2 balance of the macrophage phenotype was switched from M1 to M2 by PBM, as judged by relative staining with anti-CD86 (M1) and anti-CD206 (M2).
Martins et al looked at the effect of PBM on a model of inflammatory pain [42]. This involved injecting complete Freund's adjuvant (CFA) into the mouse paw, and produces hyperalgesia and elevated cytokine levels (TNF-α, IL-1β, IL-10). They found that LEDT (950-nm, 80 mW/cm2, 1, 2 or 4 J/cm2) applied to the plantar aspect of the right hind limb, reduced pain, increased the levels of IL-10 prevented TBARS increase in both acute and chronic phases, reduced protein carbonyl levels and increased SOD and CAT activity in the acute phase only.
Aimbire and his laboratory in Brazil have carried out several studies on the use of PBM to reduce acute lung inflammation (ALI) in various animal models. In a mouse model of lung inflammation caused either by inhalation of lipolysaccharide or intranasal administration of TNFa they analyzed the bronchoalveolar lavage fluid (BALF). PBM (660 nm, 4.5 J/cm2) was administered to the skin over the right upper bronchus 15 min after ALI induction. PBM attenuated the neutrophil influx and lowered TNFα in BALF. In alveolar macrophages, PBM increased cAMP and reduced TNFα mRNA.
They also studied a different model of ALI caused by intestinal ischemia and reperfusion (I/R), that produces an analogue of acute respiratory distress syndrome (ARDS) [78]. Rats were subjected to superior mesenteric artery occlusion (45 min) and received PBM (660 nm, 7.5 J/cm2) carried out by irradiating the rats on the skin over the right upper bronchus for 15 and 30 min, and rats were euthanized 30 min, 2, or 4 h later. PBM reduced lung edema, myeloperoxisdase activity, TNF-α and iNOS, LLLT increased IL-10 in the lungs of animals subjected to I/R.
A third animal model was related to asthma [79]. Mice were sensitized to ovalbumin (OVA), and then challenged by a single 15-min exposure to aerosolized OVA. PBM was applied as above (660 nm, 30 mW, 5.4 J). Bronchial hyper-responsiveness (as measured by dose response curves to acetylcholine) was reduced by PBM as well as reductions in eosinophils and eotaxin. PBM also diminished expression of intracellular adhesion molecule and Th2 cytokines, as well as signal transducer and activator of transduction 6 (STAT6) levels in lungs from challenged mice. Recently Rigonato-Oliveira et al. presented a study that concluded that the reduced lung inflammation and the positive effects of PBM on the airways appear to be mediated by increased secretion of the anti-inflammatory cytokine IL-10, and reduction of mucus in the airway [80].
In recent years the use of PBM as a treatment for traumatic brain injury [81,82], and other brain disorders including stroke, neurodegenerative diseases and even psychiatric disorders has increased markedly [83]. It is thought that the actions of NIR light shone on the head and penetrating into the brain are multi-factorial, but one clear effect is the anti-inflammatory action of transcranial PBM. This was shown by a series of mouse experiments conducted by Khuman et al [84]. They used the controlled cortical impact model of TBI and delivered PBM (800 nm) was applied directly to the contused parenchyma or transcranially in mice beginning 60–80min after CCI. Injured mice treated with 60J/cm2 (500 mW/cm2 × 2 min) had improved latency to the hidden platform and probe trial performance in the Morris water maze. PBM in open craniotomy mice reduced the number of activated microglia in the brain at 48 h (21.8 ± 2.3 versus 39.2 ± 4.2 IbA-1 + cells/field).
Spinal cord injury (SCI) is another promising area of central nervous system injury that could be benefited by PBM. Veronez et al [85] used a rat model of SCI involving a contusion produced by a mechanical impactor (between the ninth and tenth thoracic vertebrae), with a pressure of 150 kdyn. Three different doses of PBM (808-nm laser) were tested: 500 J/cm2, 750 J/cm2 and 1000 J/cm2 delivered daily for seven days. Functional preformance and tactile sensitivity were improved after PBM, at 1000 J/cm2. PBM at 750 and 1000 J/cm2 reduced the lesion volume and also reduced markers of inflammation (lower CD-68 protein expression).
Experimental autoimmune encephalomyelitis (EAE) is the most commonly studied animal model of multiple sclerosis (MS), a chronic autoimmune demyelinating disorder of the central nervous system. Immunomodulatory and immunosuppressive therapies currently approved for the treatment of MS slow disease progression, but do not prevent it. Lyons et al [86] studied a mouse model of EAE involving immunization with myelin oligodendrocyte glycoprotein (MOG35-55). They treated the female C57BL/6 mice with PBM (670 nm) for several days in different regimens. In addition to improved muscular function, they found down-regulation of inducible nitric oxide synthase (iNOS) gene expression in the spinal cords of mice as well as an up-regulation of the Bcl-2 anti-apoptosis gene, an increased Bcl-2:Bax ratio, and reduced apoptosis within the spinal cord of animals over the course of disease. 670 nm light therapy failed to ameliorate MOG-induced EAE in mice deficient in iNOS, confirming a role for remediation of nitrosative stress in the amelioration of MOG-induced EAE by 670 nm mediated photobiomodulation.
Yoshimura et al [87] looked at a mouse model of obesity and type 2 diabetes [87]. Four weeks old male adult C57BL/6 mice were fed a hypercaloric high-fat diet (40% calories derived from fat) for eight weeks to induce obesity and hyperglycemia. Over a period of four weeks mice were exposed to six irradiation sessions using an 843 nm LED (5.7 J cm–2, 19 mW cm–2). Non-irradiated control mice had areas of inflammation in their abdominal fat almost five times greater than the PBM group. The PBM group had significantly lower blood glucose levels 24 hours after the last session.
Amongst the many hundreds of reports of clinical applications of PBMT, we will highlight a few here, which seem to be especially relevant to inflammation, and inflammatory disorders.
Bjordal et al in Norway carried out a randomized, placebo controlled trial of PBM (904 nm, 5.4 J per point, 20 mW/cm2) for activated Achilles tendinitis [88]. In addition to clinical assessment, they used microdialysis measurement of peritendinous prostaglandin E2 concentrations. Doppler ultrasonography measurements at baseline showed minor inflammation shown by increased intra-tendinous blood flow, and a measurable resistive index. PGE2 concentrations were significantly reduced with PBM vs placebo. The pressure pain threshold also increased significantly.
Chavantes and Chammas in Brazil have studied PBM for chronic autoimmune thyroiditis. An initial pilot trial [89] used 10 applications of PBM (830 nm, 50 mW, 38–108 J/cm2), twice a week, using either the punctual technique (8 patients) or the sweep technique (7 patients). Patients required a lower dosage of levothyroxine, and showed an increased echogenicity by ultrasound. The next study [90] was a randomized, placebo-controlled trial of 43 patients with a 9-month follow-up. In addition to improved thyroid function they found reduced autoimmunity evidenced by lower thyroid peroxidase antibodies (TPOAb), and thyroglobulin antibodies (TgAb). A third study [91] used color Doppler ultrasound to show improved normal vascualrization in the thyroid parenchyma. Finally [92] they showed a statistically significant increase in serum TGF-β1 levels 30 days post-intervention in the PBM group, thus confirming the anti-inflammatory effect. Recently a long-term follow up study of these thyroiditis patients (6 years later) was presented showing that PBM was safe in the long term and demonstrated lasting benefits [93].
PBM for muscles aims to benefit athletic performance and training, to reduce delayed onset muscle soreness (DOMS), as well as to ameliorate signs of muscle damage (creatine kinase) after intense or prolonged exercise. Moreover PBM can also be used to treat frank muscle damage caused by muscle strains or trauma. The International Olympic Committee and the World Anti-Doping Agency cannot ban light therapy for athletes considering (1) the intensity is similar to sunlight, and (2) there is no forensic test for light exposure. There have been several clinical trials carried out in Brazil in athletes such as elite runners [94], volleyball players [95] and rugby players [96]. Ferraresi et al conducted a case-controlled study in a pair of identical twins [97]. They used a flexible LED array (850 nm, 75 J, 15 sec) applied to both quadriceps femoris muscles (real to one twin and sham to the other) immediately after each strength training session (3 times/wk for 12 weeks) consisting of leg press and leg extension exercises with load of 80% and 50% of the 1-repetition maximum test, respectively. PBM increased the maximal load in exercise and reduced fatigue, creatine kinase, and visual analog scale (DOMS) compared to sham. Muscle biopsies were taken before and after the training program and showed that PBM decreased inflammatory markers such as interleukin 1β and muscle atrophy (myostatin). Protein synthesis (mammalian target of rapamycin) and oxidative stress defense (SOD2, mitochondrial superoxide dismutase) were up-regulated.
Psoriasis is a chronic autoimmune skin disease. Psoriasis is characterized by the abnormally excessive and rapid growth of keratinocytes (instead of being replaced every 28–30 days as in normal skin, in psoriatic skin they are replaced every 3–5 days). This hyperproliferation is caused by an inflammatory cascade in the dermis involving dendritic cells, macrophages, and T cells secreting TNF-α, IL-1β, IL-6, IL-17, IL-22, and IL-36γ [98]. PBM has been used for psoriasis because of its anti-inflammatory effects, which is a different approach from UV phototherapy which tends to kill circulating T-cells. Ablon [99] tested PBM using LEDs (830 nm, 60 J/cm2 and 633 nm, 126 J/cm2) in two 20-min sessions over 4 or 5 weeks, with 48 h between sessions in 9 patients with chronic treatment-resistant psoriasis. Clearance rates at the end of the follow-up period ranged from 60% to 100%. Satisfaction was universally very high.
Choi et al [100] tested PBM in case report of a patient with another inflammatory skin disease called acrodermatitis continua, who also had a 10-yr history of plaque psoriasis on her knees and elbows. As she was pregnant and not suited for pharmacological therapy, she received treatment with PBM (broad-band polarized light, 480–3,400 nm, 10 J/cm2). In two weeks (after only 4 treatments), the clinical resolution was impressive and no pustules were found. Topical methylprednisolone aceponate steroid cream was switched to a moisturizer, and she was treated twice or once a week with PBM until a healthy baby was delivered.
As can be seen from the animal studies section, arthritis is one of the most important clinical indications for PBM [101,102]. The two most common forms of arthritis are osteoarthritis (degenerative joint disease that mostly affects the fingers, knees, and hips) and rheumatoid arthritis (autoimmune joint inflammation that often affects the hands and feet). Osteoarthritis (OA) affects more than 3.8% of the population while rheumatoid arthritis (RA) affects about 0.24%. Both types have been successfully treated with PBM. Cochrane systematic reviews found for good evidence for its effectiveness in RA [103], and some evidence in the case of OA [104]. Most clinical studies have used pain scales and range of movement scores to test the effectiveness, rather than measures of inflammation which are difficult to carry out in human subjects.
Barabas and coworkers [105] made an attempt by testing PBM on ex vivo samples of synovial tissue surgically removed from patients receiving knee joint replacement. Synovial membrane samples received exposure to PBM (810 nm, 448 mW, 25 J/cm2, 1 cm2 area). PBM caused an increase in mitochondrial heat shock protein 1 60 kD, and decreases in calpain small subunit 1, tubulin alpha-1C, beta 2, vimentin variant 3, annexin A1, annexin A5, cofilin 1, transgelin, and collagen type Ⅵ alpha 2 chain precursor all significantly decreased compared to the control
Alopecia areata (AA) is one of the three common types of hair loss, the other two being androgenetic alopecia (AGA, male pattern baldness) and chemotherapy induced alopecia. AA is a common autoimmune disease resulting from damage caused to the hair follicles (HFs) by T cells. Evidence of autoantibodies to anagen stage HF structures is found in affected humans and experimental mouse models. Biopsy specimens from affected individuals demonstrate a characteristic peri-and intrafollicular inflammatory infiltrate around anagen-stage HFs consisting of activated CD4 and CD8 T lymphocytes [106]. PBM is an excellent treatment for hair loss in general and AGA in particular [107,108]. Yamazaki et al [109] reported the use of the "Super-Lizer" delivering linear-polarized light between 600–1600 nm at a power of 1.26 W to the areas of hair loss on the scalp (4-s pulses delivered at 1-s intervals for 3 min every 1 or 2 weeks until hair growth was observed). Regrowth of vellus hairs was achieved on more than 50% ofthe involved areas in all 15 cases. The frequency of irradiation until regrowth ranged from one to 14 times and the duration of SL treatment was 2 weeks to 5 months.
The clinical applications of PBM have been increasing apace in recent years. The recent adoption of inexpensive large area LED arrays, that have replaced costly, small area laser beams with a risk of eye damage, has accelerated this increase in popularity. Advances in understanding of PBM mechanisms of action at a molecular and cellular level, have provided a scientific rationale for its use for multiple diseases. Many patients have become disillusioned with traditional pharmaceutical approaches to a range of chronic conditions, with their accompanying distressing side-effects and have turned to complementary and alternative medicine for more natural remedies. PBM has an almost complete lack of reported adverse effects, provided the parameters are understood at least at a basic level. The remarkable range of medical benefits provided by PBM, has led some to suggest that it may be "too good to be true". However one of the most general benefits of PBM that has recently emerged, is its pronounced anti-inflammatory effects. While the exact cellular signaling pathways responsible for this anti-inflammatory action are not yet completely understood, it is becoming clear that both local and systemic mechanisms are operating. The local reduction of edema, and reductions in markers of oxidative stress and pro-inflammatory cytokines are well established. However there also appears to be a systemic effect whereby light delivered to the body, can positively benefit distant tissues and organs.
There is a lot of scope for further work on PBM and inflammation. The intriguing benefits of PBM on some autoimmune diseases, suggests that this area may present a fertile area for researchers. There may be some overlap between the ability of PBM to activate and mobilize stem cells and progenitor cells, and its anti-inflammatory action, considering that one of the main benefits of exogenous stem cell therapy has been found to be its anti-inflammatory effect. The versatile benefits of PBM on the brain and the central nervous system, encourages further study of its ability to reduce neuroinflammation. Chronic diseases of the modern age involving systemic inflammation such as type Ⅱ diabetes, obesity, Alzheimer's disease, cardiovascular disease and endothelial dysfunction are again worth investigating in the context of PBM.
MRH was supported by US NIH grants R01AI050875 and R21AI121700.
The author declares no conflict of interest in this paper.
[1] |
A. Wibisono, A. Wilson, M. Jordan, A variational perspective on accelerated methods in optimization, Proc. Natl. Acad. Sci., 113 (2016), E7351–E7358. https://doi.org/10.1073/pnas.1614734113 doi: 10.1073/pnas.1614734113
![]() |
[2] |
V. Duruisseaux, M. Leok, A variational formulation of accelerated optimization on Riemannian manifolds, SIAM J. Math. Data Sci., 4 (2022), 649–674. https://doi.org/10.1137/21M1395648 doi: 10.1137/21M1395648
![]() |
[3] |
V. Duruisseaux, J. Schmitt, M. Leok, Adaptive Hamiltonian variational integrators and applications to symplectic accelerated optimization, SIAM J. Sci. Comput., 43 (2021), A2949–A2980. https://doi.org/10.1137/20M1383835 doi: 10.1137/20M1383835
![]() |
[4] | Y. Nesterov, A method of solving a convex programming problem with convergence rate . Sov. Math. Dokl., 27 (1983), 372–376. |
[5] | Y. Nesterov, Introductory Lectures on Convex Optimization: A Basic Course, volume 87 of Applied Optimization, Kluwer Academic Publishers, Boston, MA, 2004. |
[6] | W. Su, S. Boyd, E. Candes, A differential equation for modeling Nesterov's Accelerated Gradient method: theory and insights. J. Mach. Learn. Res., 17 (2016), 1–43. |
[7] | M. Betancourt, M. I. Jordan, A. Wilson, On symplectic optimization. 2018. https://doi.org/10.48550/arXiv.1802.03653 |
[8] | C. M. Campos, A. Mahillo, D. Martín de Diego, A discrete variational derivation of accelerated methods in optimization. 2021. https://doi.org/10.48550/arXiv.2106.02700 |
[9] |
G. França, M. I. Jordan, R. Vidal, On dissipative symplectic integration with applications to gradient-based optimization. J. Stat. Mech., 2021 (2021), 043402. https://doi.org/10.1088/1742-5468/abf5d4 doi: 10.1088/1742-5468/abf5d4
![]() |
[10] | G. França, A. Barp, M. Girolami, M. I. Jordan, Optimization on manifolds: A symplectic approach, 07 2021. https://doi.org/10.48550/arXiv.2107.11231 |
[11] | M. I. Jordan, Dynamical, symplectic and stochastic perspectives on gradient-based optimization, In Proceedings of the International Congress of Mathematicians (ICM 2018), 523–549. https://doi.org/10.1142/9789813272880_0022 |
[12] | E. Hairer, C. Lubich, G. Wanner, Geometric Numerical Integration, volume 31 of Springer Series in Computational Mathematics, Springer-Verlag, Berlin, second edition, 2006. |
[13] |
J. P. Calvo, J. M. Sanz-Serna, The development of variable-step symplectic integrators, with application to the two-body problem, SIAM J. Sci. Comp., 14 (1993), 936–952. https://doi.org/10.1137/0914057 doi: 10.1137/0914057
![]() |
[14] |
B. Gladman, M. Duncan, J. Candy, Symplectic integrators for long-time integrations in celestial mechanics. Celestial Mech. Dyn. Astr., 52 (1991), 221–240. https://doi.org/10.1007/BF00048485 doi: 10.1007/BF00048485
![]() |
[15] |
E. Hairer, Variable time step integration with symplectic methods, Appl. Numer. Math., 25 (1997), 219–227. https://doi.org/10.1016/S0168-9274(97)00061-5 doi: 10.1016/S0168-9274(97)00061-5
![]() |
[16] |
K. Zare, V. G. Szebehely, Time transformations in the extended phase-space. Celestial Mech., 11 (1975), 469–482. https://doi.org/10.1007/BF01650285 doi: 10.1007/BF01650285
![]() |
[17] |
J. Hall, M. Leok, Spectral Variational Integrators, Numer. Math., 130 (2015), 681–740. https://doi.org/10.1007/s00211-014-0679-0 doi: 10.1007/s00211-014-0679-0
![]() |
[18] |
J. E. Marsden, M. West, Discrete mechanics and variational integrators. Acta Numer., 10 (2001), 357–514. https://doi.org/10.1017/S096249290100006X doi: 10.1017/S096249290100006X
![]() |
[19] | T. Lee, M. Tao, M. Leok, Variational symplectic accelerated optimization on Lie groups. 2021. |
[20] | M. Tao, T. Ohsawa, Variational optimization on Lie groups, with examples of leading (generalized) eigenvalue problems. In Proceedings of the 23rd International AISTATS Conference, volume 108 of PMLR, 2020. |
[21] | F. Alimisis, A. Orvieto, G. Bécigneul, A. Lucchi, A continuous-time perspective for modeling acceleration in Riemannian optimization. In Proceedings of the 23rd International AISTATS Conference, volume 108 of PMLR, 1297–1307, 2020. |
[22] |
V. Duruisseaux, M. Leok, Accelerated optimization on Riemannian manifolds via discrete constrained variational integrators. J. Nonlinear Sci., 32 (2022). https://doi.org/10.1007/s00332-022-09795-9 doi: 10.1007/s00332-022-09795-9
![]() |
[23] | V. Duruisseaux, M. Leok, Accelerated optimization on Riemannian manifolds via projected variational integrators. 2022. https://doi.org/10.48550/arXiv.2201.02904 |
[24] |
J. Nash, The imbedding problem for Riemannian manifolds, Ann. Math., 63 (1956), 20–63. https://doi.org/10.2307/1969989 doi: 10.2307/1969989
![]() |
[25] |
H. Whitney, The self-intersections of a smooth -manifold in -space, Ann. Math., 45 (1944), 220–246. https://doi.org/10.2307/1969265 doi: 10.2307/1969265
![]() |
[26] |
H. Whitney, The singularities of a smooth -manifold in -space, Ann. Math., 45 (1944), 247–293. https://doi.org/10.2307/1969266 doi: 10.2307/1969266
![]() |
[27] |
N. Bou-Rabee, J. E. Marsden, Hamilton–Pontryagin integrators on Lie groups part Ⅰ: Introduction and structure-preserving properties, Found. Comput. Math., 9 (2009), 197–219. https://doi.org/10.1007/s10208-008-9030-4 doi: 10.1007/s10208-008-9030-4
![]() |
[28] |
I. I. Hussein, M. Leok, A. K. Sanyal, A. M. Bloch, A discrete variational integrator for optimal control problems on SO(3). Proceedings of the 45th IEEE Conference on Decision and Control, 6636–6641, 2006. https://doi.org/10.1109/CDC.2006.377818 doi: 10.1109/CDC.2006.377818
![]() |
[29] | T. Lee, Computational geometric mechanics and control of rigid bodies. Ph.D. dissertation, University of Michigan, 2008. |
[30] |
T. Lee, M. Leok, N. H. McClamroch, Lie group variational integrators for the full body problem, Comput. Methods Appl. Mech. Engrg., 196 (2007), 2907–2924. https://doi.org/10.1016/j.cma.2007.01.017 doi: 10.1016/j.cma.2007.01.017
![]() |
[31] |
T. Lee, M. Leok, N. H. McClamroch, Lie group variational integrators for the full body problem in orbital mechanics, Celestial Mech. Dyn. Astr., 98 (2007), 121–144. https://doi.org/10.1007/s10569-007-9073-x doi: 10.1007/s10569-007-9073-x
![]() |
[32] |
T. Lee, N. H. McClamroch, M. Leok, A Lie group variational integrator for the attitude dynamics of a rigid body with applications to the 3d pendulum, Proceedings of 2005 IEEE Conference on Control Applications. (CCA 2005), 962–967. https://doi.org/10.1109/CCA.2005.1507254 doi: 10.1109/CCA.2005.1507254
![]() |
[33] | M. Leok, Generalized Galerkin variational integrators: Lie group, multiscale, and pseudospectral methods, (preprint, arXiv: math.NA/0508360), 2004. |
[34] | N. Nordkvist, A. K. Sanyal, A Lie group variational integrator for rigid body motion in SE(3) with applications to underwater vehicle dynamics, In 49th IEEE Conference on Decision and Control (CDC), 5414–5419, 2010. https://doi.org/10.1109/CDC.2010.5717622 |
[35] |
T. Lee, M. Leok, N. H. McClamroch, Lagrangian mechanics and variational integrators on two-spheres, Int. J. Numer. Methods Eng., 79 (2009), 1147–1174. https://doi.org/10.1002/nme.2603 doi: 10.1002/nme.2603
![]() |
[36] | J. E. Marsden, T. S. Ratiu, Introduction to mechanics and symmetry, volume 17 of Texts in Applied Mathematics. Springer-Verlag, New York, second edition, 1999. |
[37] |
S. Lall, M. West, Discrete variational Hamiltonian mechanics, J. Phys. A, 39 (2006), 5509–5519. https://doi.org/10.1088/0305-4470/39/19/S11 doi: 10.1088/0305-4470/39/19/S11
![]() |
[38] |
M. Leok, J. Zhang, Discrete Hamiltonian variational integrators, IMA J. Numer. Anal., 31 (2011), 1497–1532. https://doi.org/10.1093/imanum/drq027 doi: 10.1093/imanum/drq027
![]() |
[39] |
J. M. Schmitt, M. Leok, Properties of Hamiltonian variational integrators, IMA J. Numer. Anal., 38 (2017), 377–398. https://doi.org/10.1093/imanum/drx010 doi: 10.1093/imanum/drx010
![]() |
[40] |
M. Leok, T. Shingel, Prolongation-collocation variational integrators, IMA J. Numer. Anal., 32 (2012), 1194–1216. https://doi.org/10.1093/imanum/drr042 doi: 10.1093/imanum/drr042
![]() |
[41] |
J. M. Schmitt, T. Shingel, M. Leok, Lagrangian and Hamiltonian Taylor variational integrators, BIT Numer. Math., 58 (2018), 457–488. https://doi.org/10.1007/s10543-017-0690-9 doi: 10.1007/s10543-017-0690-9
![]() |
[42] |
M. Leok, T. Ohsawa, Variational and geometric structures of discrete Dirac mechanics, Found. Comput. Math., 11 (2011), 529–562. https://doi.org/10.1007/s10208-011-9096-2 doi: 10.1007/s10208-011-9096-2
![]() |
[43] |
H. Yoshimura, J. E. Marsden, Dirac structures in Lagrangian mechanics Part Ⅰ: Implicit Lagrangian systems, J. Geom. Phys., 57 (2006), 133–156. https://doi.org/10.1016/j.geomphys.2006.02.009 doi: 10.1016/j.geomphys.2006.02.009
![]() |
[44] |
H. Yoshimura, J. E. Marsden, Dirac structures in Lagrangian mechanics Part Ⅱ: Variational structures, J. Geom. Phys., 57 (2006), 209–250. https://doi.org/10.1016/j.geomphys.2006.02.012 doi: 10.1016/j.geomphys.2006.02.012
![]() |
[45] | D. Kingma, J. Ba, Adam: A method for stochastic optimization, International Conference on Learning Representations, 12 2014. https://doi.org/10.48550/arXiv.1412.6980 |
[46] | J. Duchi, E. Hazan, Y. Singer, Adaptive subgradient methods for online learning and stochastic optimization, J. Mach. Learn. Res., 12 (2011), 2121–2159. |
[47] | T. Tieleman, G. Hinton, Coursera: Neural Networks for Machine Learning - Lecture 6.5: RMSprop, 2012. |
[48] | P. A. Absil, R. Mahony, R. Sepulchre, Optimization Algorithms on Matrix Manifolds, Princeton University Press, Princeton, NJ, 2008. https://doi.org/10.1515/9781400830244 |
[49] | N. Boumal, An introduction to optimization on smooth manifolds, 2020. |
[50] | J. M. Lee, Introduction to Riemannian Manifolds, volume 176 of Graduate Texts in Mathematics, Springer, Cham, second edition, 2018. |
[51] | J. Jost, Riemannian Geometry and Geometric Analysis, Universitext. Springer, Cham, 7th edition, 2017. |
[52] | T. Lee, M. Leok, N. H. McClamroch, Global Formulations of Lagrangian and Hamiltonian Dynamics on Manifolds: A Geometric Approach to Modeling and Analysis, Interaction of Mechanics and Mathematics. Springer International Publishing, 2017. |
[53] | J. Gallier, J. Quaintance, Differential Geometry and Lie Groups: A Computational Perspective, Geometry and Computing. Springer International Publishing, 2020. |
[54] |
E. Celledoni, H. Marthinsen, B. Owren, An introduction to Lie group integrators – basics, new developments and applications, J. Comput. Phys., 257 (2014), 1040–1061. https://doi.org/10.1016/j.jcp.2012.12.031 doi: 10.1016/j.jcp.2012.12.031
![]() |
[55] |
E. Celledoni, E. Çokaj, A. Leone, D. Murari, B. Owren, Lie group integrators for mechanical systems, Int. J. Comput. Math., 99 (2022), 58–88. https://doi.org/10.1080/00207160.2021.1966772 doi: 10.1080/00207160.2021.1966772
![]() |
[56] |
S. H. Christiansen, H. Z. Munthe-Kaas, B. Owren, Topics in structure-preserving discretization, Acta Numer., 20 (2011), 1–119. https://doi.org/10.1017/S096249291100002X doi: 10.1017/S096249291100002X
![]() |
[57] | A. Iserles, H. Z. Munthe-Kaas, S. P. Nørsett, A. Zanna, Lie group methods, Acta Numer., 9 (2000), 215–365. https://doi.org/10.1017/S0962492900002154 |
[58] |
P. E. Crouch, R. L. Grossman, Numerical integration of ordinary differential equations on manifolds, J. Nonlinear Sci., 3 (1993), 1–33. https://doi.org/10.1007/BF02429858 doi: 10.1007/BF02429858
![]() |
[59] |
D. Lewis, J. C. Simo, Conserving algorithms for the dynamics of Hamiltonian systems on Lie groups, J. Nonlinear Sci., 4 (1994), 253–299. https://doi.org/10.1007/BF02430634 doi: 10.1007/BF02430634
![]() |
[60] |
F. Casas, B. Owren, Cost efficient Lie group integrators in the RKMK class, BIT, 43 (2003), 723–742. https://doi.org/10.1023/B:BITN.0000009959.29287.d4 doi: 10.1023/B:BITN.0000009959.29287.d4
![]() |
[61] |
H. Z. Munthe-Kaas, Lie-Butcher theory for Runge-Kutta methods, BIT, 35 (1995), 572–587. https://doi.org/10.1007/BF01739828 doi: 10.1007/BF01739828
![]() |
[62] |
H. Z. Munthe-Kaas, Runge-Kutta methods on Lie groups, Bit Numer. Math., 38 (1998), 92–111. https://doi.org/10.1007/BF02510919 doi: 10.1007/BF02510919
![]() |
[63] |
H. Z. Munthe-Kaas, High order Runge-Kutta methods on manifolds, Appl. Numer. Math., 29 (1999), 115–127. https://doi.org/10.1016/S0168-9274(98)00030-0 doi: 10.1016/S0168-9274(98)00030-0
![]() |
[64] |
S. Blanes, F. Casas, J. A. Oteo, J. Ros, The Magnus expansion and some of its applications, Phys. Rep., 470 (2008), 151–238. https://doi.org/10.1016/j.physrep.2008.11.001 doi: 10.1016/j.physrep.2008.11.001
![]() |
[65] |
A. Iserles, S. P. Nørsett, On the solution of linear differential equations in Lie groups, Phil. Trans. R. Soc. A, 357 (1999), 983–1019. https://doi.org/10.1098/rsta.1999.0362 doi: 10.1098/rsta.1999.0362
![]() |
[66] |
A. Zanna, Collocation and relaxed collocation for the Fer and the Magnus expansions, SIAM J. Numer. Anal., 36 (1999), 1145–1182. https://doi.org/10.1137/S0036142997326616 doi: 10.1137/S0036142997326616
![]() |
[67] |
E. Celledoni, A. Marthinsen, B. Owren, Commutator-free Lie group methods, Future Gener. Comput. Syst., 19 (2003), 341–352. https://doi.org/10.1016/S0167-739X(02)00161-9 doi: 10.1016/S0167-739X(02)00161-9
![]() |
[68] |
G. Wahba, A Least Squares Estimate of Satellite Attitude, SIAM Rev., 7 (1965), 409. https://doi.org/10.1137/1007077 doi: 10.1137/1007077
![]() |
[69] | V. Duruisseaux, M. Leok, Practical perspectives on symplectic accelerated optimization, 2022. https://doi.org/10.48550/arXiv.2207.11460 |
![]() |
![]() |
1. | Lucas Andreo, Beatriz G. Ribeiro, Agnelo N. Alves, Andréia S.A. Martinelli, Carla B. Soldera, Anna Carolina R.T. Horliana, Sandra Kalil Bussadori, Kristianne P.S. Fernandes, Raquel A. Mesquita‐Ferrari, Effects of Photobiomodulation with Low‐level Laser Therapy on Muscle Repair Following a Peripheral Nerve Injury in Wistar Rats, 2020, 96, 0031-8655, 1124, 10.1111/php.13255 | |
2. | Jociane Schardong, Mariana Falster, Isadora Rebolho Sisto, Ana Paula Oliveira Barbosa, Tatiana Coser Normann, Kellen Sábio de Souza, Gabriela Jaroceski, Camila Bassani Bozzetto, Bruno Manfredini Baroni, Rodrigo Della Méa Plentz, Photobiomodulation therapy increases functional capacity of patients with chronic kidney failure: randomized controlled trial, 2021, 36, 0268-8921, 119, 10.1007/s10103-020-03020-3 | |
3. | Chaiyarerk Homsirikamol, Saroj Suvanasuthi, Kwanchanok Viravaidya-Pasuwat, Validation of uniformity-optimized irradiance distribution on a well-plate platform from a light-emitting-diode array, 2020, 59, 1559-128X, 6168, 10.1364/AO.392126 | |
4. | Linda L. Chao, Cody Barlow, Mahta Karimpoor, Lew Lim, Changes in Brain Function and Structure After Self-Administered Home Photobiomodulation Treatment in a Concussion Case, 2020, 11, 1664-2295, 10.3389/fneur.2020.00952 | |
5. | Reem Hanna, Snehal Dalvi, Tudor Sălăgean, Ioana Roxana Bordea, Stefano Benedicenti, Phototherapy as a Rational Antioxidant Treatment Modality in COVID-19 Management; New Concept and Strategic Approach: Critical Review, 2020, 9, 2076-3921, 875, 10.3390/antiox9090875 | |
6. | Mariana A Vetrici, Soheila Mokmeli, Andrew R Bohm, Monica Monici, Scott A Sigman, Evaluation of Adjunctive Photobiomodulation (PBMT) for COVID-19 Pneumonia via Clinical Status and Pulmonary Severity Indices in a Preliminary Trial, 2021, Volume 14, 1178-7031, 965, 10.2147/JIR.S301625 | |
7. | Vladimir Heiskanen, Morgan Pfiffner, Timo Partonen, Sunlight and health: shifting the focus from vitamin D3 to photobiomodulation by red and near-infrared light, 2020, 61, 15681637, 101089, 10.1016/j.arr.2020.101089 | |
8. | W. R. Beasi, L. V. Toffoli, G. G. Pelosi, M. V. M. Gomes, L. F. Verissimo, M. R. Stocco, L. C. Mantoani, L. P. Maia, R. A. C. Andraus, Effects of photobiomodulation and swimming on gene expression in rats with the tibialis anterior muscle injury, 2020, 0268-8921, 10.1007/s10103-020-03168-y | |
9. | Shanshan Wei, Xiaotong Ren, Yuexin Wang, Yilin Chou, Xuemin Li, Therapeutic Effect of Intense Pulsed Light (IPL) Combined with Meibomian Gland Expression (MGX) on Meibomian Gland Dysfunction (MGD), 2020, 2020, 2090-004X, 1, 10.1155/2020/3684963 | |
10. | Hyeyoon Goo, Hoon Kim, Jin-Chul Ahn, Kyong Jin Cho, Effects of Low-level Light Therapy at 740 nm on Dry Eye Disease In Vivo, 2019, 8, 2287-8300, 50, 10.25289/ML.2019.8.2.50 | |
11. | Lilach Gavish, Dan Gilon, Ronen Beeri, Ayelet Zuckerman, Dean Nachman, S. David Gertz, Photobiomodulation and estrogen stabilize mitochondrial membrane potential in angiotensin– II challenged porcine aortic smooth muscle cells , 2021, 14, 1864-063X, 10.1002/jbio.202000329 | |
12. | Luan dos Santos Mendes-Costa, Vanessa Garcia de Lima, Maria Paula Ribeiro Barbosa, Ludmila Evangelista dos Santos, Suélia de Siqueira Rodrigues Fleury Rosa, José Carlos Tatmatsu-Rocha, Photobiomodulation: systematic review and meta-analysis of the most used parameters in the resolution diabetic foot ulcers, 2020, 0268-8921, 10.1007/s10103-020-03192-y | |
13. | Asheesh Gupta, Gaurav K. Keshri, Anju Yadav, Non-thermal Therapeutic Applications of Light, 2018, 88, 0369-8203, 473, 10.1007/s40010-018-0521-7 | |
14. | Caiyun Meng, Qing Xia, Hao Wu, He Huang, Hailiang Liu, Yujun Li, Fengmin Zhang, Wuqi Song, Photobiomodulation with 630-nm LED radiation inhibits the proliferation of human synoviocyte MH7A cells possibly via TRPV4/PI3K/AKT/mTOR signaling pathway, 2020, 35, 0268-8921, 1927, 10.1007/s10103-020-02977-5 | |
15. | Wen-Dien Chang, Jih-Huah Wu, Nai-Jen Chang, Chia-Lun Lee, Shuya Chen, Effects of Laser Acupuncture on Delayed Onset Muscle Soreness of the Biceps Brachii Muscle: A Randomized Controlled Trial, 2019, 2019, 1741-427X, 1, 10.1155/2019/6568976 | |
16. | Jean-Paul Rocca, Meng Zhao, Carlo Fornaini, Lixin Tan, Zengyi Zhao, Elisabetta Merigo, Effect of laser irradiation on aphthae pain management: A four different wavelengths comparison, 2018, 189, 10111344, 1, 10.1016/j.jphotobiol.2018.09.016 | |
17. | Reza Zomorrodi, Genane Loheswaran, Lew Lim, 2019, 9780128153055, 419, 10.1016/B978-0-12-815305-5.00030-0 | |
18. | J. Taradaj, K. Rajfur, J. Rajfur, K. Ptaszkowski, L. Ptaszkowska, M. Sopel, J. Rosińczuk, R. Dymarek, Effect of laser treatment on postural control parameters in patients with chronic nonspecific low back pain: a randomized placebo-controlled trial, 2019, 52, 1414-431X, 10.1590/1414-431x20198474 | |
19. | Fereshteh Safian, Marefat Ghaffari Novin, Hamid Nazarian, Zahra Shams Mofarahe, Mohammad-Amin Abdollahifar, Vahid Jajarmi, Sareh Karimi, Mahsa Kazemi, Sufan Chien, Mohammad Bayat, Photobiomodulation preconditioned human semen protects sperm cells against detrimental effects of cryopreservation, 2021, 98, 00112240, 239, 10.1016/j.cryobiol.2020.09.005 | |
20. | Sara Pourshahidi, Elaheh Ghasem Zadeh Hoseini, Shiva Shirazian, Hooman Ebrahimi, Nasim Chiniforush, Mojgan Alaeddini, Shahroo Etemad‐Moghadam, Rui Amaral Mendes, The Effect of Laser Bio‐modulation on Dysplastic Lesions, an Animal Study, 2021, 0031-8655, 10.1111/php.13400 | |
21. | Nadhia H. C. Souza, Raquel A. Mesquita‐Ferrari, Maria Fernanda S. D. Rodrigues, Daniela F. T. Silva, Beatriz G. Ribeiro, Agnelo N. Alves, Mónica P. Garcia, Fábio D. Nunes, Evaldo M. Silva Junior, Cristiane M. França, Sandra K. Bussadori, Kristianne P. S. Fernandes, Photobiomodulation and different macrophages phenotypes during muscle tissue repair, 2018, 22, 1582-1838, 4922, 10.1111/jcmm.13757 | |
22. | Carlos Alberto Tenis, Manoela Domingues Martins, Marcela Leticia Leal Gonçalves, Daniela de Fátima Teixeira da Silva, João Júlio da Cunha Filho, Marco Antonio Trevizani Martins, Raquel Agnelli Mesquita-Ferrari, Sandra Kalil Bussadori, Kristianne Porta Santos Fernandes, Efficacy of light-emitting diode (LED) photobiomodulation in pain management, facial edema, trismus, and quality of life after extraction of retained lower third molars, 2018, 97, 0025-7974, e12264, 10.1097/MD.0000000000012264 | |
23. | Patrícia Gabrielli Vassão, Gabriel Sobrinho Baldini, Kamila Verlene S.G. Vieira, Ana Beatriz Balão, Carlos Eduardo Pinfildi, Raquel Munhoz da Silveira Campos, Helga Tatiana Tucci, Ana Claudia Muniz Renno, Acute Photobiomodulation Effects Through a Cluster Device on Skeletal Muscle Fatigue of Biceps Brachii in Young and Healthy Males: A Randomized Double-Blind Session, 2020, 38, 2578-5478, 773, 10.1089/photob.2019.4786 | |
24. | Mateus Rossato, Rodolfo André Dellagrana, Raphael Luiz Sakugawa, Bruno Manfredini Baroni, Fernando Diefenthaeler, Dose–Response Effect of Photobiomodulation Therapy on Muscle Performance and Fatigue During a Multiple-Set Knee Extension Exercise: A Randomized, Crossover, Double-Blind Placebo-Controlled Trial, 2020, 38, 2578-5478, 758, 10.1089/photob.2020.4820 | |
25. | Mariana Rodrigues, Ramon B. Cardoso, Heloyse U. Kuriki, Alexandre M. Marcolino, Elaine Caldeira Oliveira Guirro, Rafael I. Barbosa, Photobiomodulation Decreases Hyperalgesia in Complex Regional Pain Syndrome: An Experimental Mouse Model Subjected to Nicotine, 2020, 52, 0196-8092, 890, 10.1002/lsm.23240 | |
26. | Deirdre Edge, Mikkel Schødt, Michael Canova Engelbrecht Nielsen, 2020, Chapter 37, 978-3-030-45350-3, 387, 10.1007/978-3-030-45351-0_37 | |
27. | Larissa Alexsandra da Silva Neto Trajano, Luiz Philippe da Silva Sergio, Ana Carolina Stumbo, Andre Luiz Mencalha, Adenilson de Souza da Fonseca, Low power lasers on genomic stability, 2018, 180, 10111344, 186, 10.1016/j.jphotobiol.2018.02.010 | |
28. | Vanessa Christina Santos Pavesi, Manoela Domingues Martins, Fábio Luiz Coracin, Aline Silva Sousa, Benedito Jorge Pereira, Renato Araújo Prates, Juliana Freitas da Silva, Marcela Leticia Leal Gonçalves, Cicero Dayves Silva Bezerra, Sandra Kalil Bussadori, Maria Lucia Zarvos Varellis, Alessandro Melo Deana, Effects of photobiomodulation in salivary glands of chronic kidney disease patients on hemodialysis, 2021, 0268-8921, 10.1007/s10103-020-03158-0 | |
29. | Jennifer A. Brown, Julia Tomlinson, Rehabilitation of the Canine Forelimb, 2021, 51, 01955616, 401, 10.1016/j.cvsm.2020.12.005 | |
30. | Gianni Betti, Costanza Consolandi, Robert G. Eccles, The Relationship between Investor Materiality and the Sustainable Development Goals, 2018, 1556-5068, 10.2139/ssrn.3163044 | |
31. | Jonathan Stone, John Mitrofanis, Daniel M. Johnstone, Benedetto Falsini, Silvia Bisti, Paul Adam, Arturo Bravo Nuevo, Mindy George-Weinstein, Rebecca Mason, Janis Eells, Acquired Resilience: An Evolved System of Tissue Protection in Mammals, 2018, 16, 1559-3258, 155932581880342, 10.1177/1559325818803428 | |
32. | Andrea Marchegiani, Andrea Spaterna, Matteo Cerquetella, Current Applications and Future Perspectives of Fluorescence Light Energy Biomodulation in Veterinary Medicine, 2021, 8, 2306-7381, 20, 10.3390/vetsci8020020 | |
33. | Wei-Ting Liao, Chih-Hsing Hung, Shih-Shin Liang, Sebastian Yu, Jian-He Lu, Chih-Hung Lee, Chee-Yin Chai, Hsin-Su Yu, Anti-inflammatory effects induced by near-infrared light irradiation via M2 macrophage polarization, 2021, 0022202X, 10.1016/j.jid.2020.11.035 | |
34. | Jinatta Jittiwat, Baihui Point Laser Acupuncture Ameliorates Cognitive Impairment, Motor Deficit, and Neuronal Loss Partly via Antioxidant and Anti-Inflammatory Effects in an Animal Model of Focal Ischemic Stroke, 2019, 2019, 1741-427X, 1, 10.1155/2019/1204709 | |
35. | Zeqing Chen, Haokuan Qin, Shangfei Lin, Zhicheng Lu, Xuewei Fan, Xuwen Liu, Muqing Liu, Comparative transcriptome analysis of gene expression patterns on B16F10 melanoma cells under Photobiomodulation of different light modes, 2021, 216, 10111344, 112127, 10.1016/j.jphotobiol.2021.112127 | |
36. | Maria Rosa Antognazza, Ilaria Abdel Aziz, Francesco Lodola, Use of Exogenous and Endogenous Photomediators as Efficient ROS Modulation Tools: Results and Perspectives for Therapeutic Purposes, 2019, 2019, 1942-0900, 1, 10.1155/2019/2867516 | |
37. | Zhuowen Liang, Tao Lei, Shuang Wang, Xiaoshuang Zuo, Kun Li, Jiwei Song, Jiakai Sun, Jiawei Zhang, Qiao Zheng, Xiaowei Kang, Yangguang Ma, Xueyu Hu, Tan Ding, Zhe Wang, Photobiomodulation by diffusing optical fiber on spinal cord: A feasibility study in piglet model, 2020, 13, 1864-063X, 10.1002/jbio.201960022 | |
38. | Qi Shen, Lei Liu, Xiaotong Gu, Da Xing, Photobiomodulation suppresses JNK3 by activation of ERK/MKP7 to attenuate AMPA receptor endocytosis in Alzheimer's disease, 2021, 20, 1474-9718, 10.1111/acel.13289 | |
39. | Luodan Yang, Yan Dong, Chongyun Wu, Yong Li, Yichen Guo, Baocheng Yang, Xuemei Zong, Michael R. Hamblin, Timon C.‐Y. Liu, Quanguang Zhang, Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia‐ischemia, 2019, 12, 1864-063X, 10.1002/jbio.201800359 | |
40. | Angela Maria Paiva Magri, Júlia Risso Parisi, Ana Laura Martins Andrade, Ana Claudia Muniz Rennó, Bone substitutes and photobiomodulation in bone regeneration: A systematic review in animal experimental studies, 2021, 1549-3296, 10.1002/jbm.a.37170 | |
41. | John C. Castel, Dawn Castel, Tony Wei, Michael R. Hamblin, James D. Carroll, Praveen Arany, 2019, Novel technology platform for PBM delivery using printed LEDs, 9781510623644, 13, 10.1117/12.2508729 | |
42. | Ruan Felipe Ferreira Tomé, Diego Filipe Bezerra Silva, Carlus Alberto Oliveira dos Santos, Gabriella de Vasconcelos Neves, Ana Karina Almeida Rolim, Daliana Queiroga de Castro Gomes, ILIB (intravascular laser irradiation of blood) as an adjuvant therapy in the treatment of patients with chronic systemic diseases—an integrative literature review, 2020, 35, 0268-8921, 1899, 10.1007/s10103-020-03100-4 | |
43. | Sogol Yousefi, Vida Hojati, Farinaz Nasirinezhad, Fatemeh Ramezani, Atousa Janzadeh, Gholamhasan Vaezi, The Effect of Four Weeks of Low-Level Laser Radiation (660 nm) on Movement Recovery and Fibroblasts Invasion, 2019, 6, 2322-3944, 10.5812/ans.87225 | |
44. | Brian Bicknell, Ann Liebert, Daniel Johnstone, Hosen Kiat, Photobiomodulation of the microbiome: implications for metabolic and inflammatory diseases, 2019, 34, 0268-8921, 317, 10.1007/s10103-018-2594-6 | |
45. | Carlos Alberto Ocon, Solange Almeida dos Santos, Jheniphe Rocha Caires, Marcelo Ferreira Duarte de Oliveira, Andrey Jorge Serra, Ernesto Cesar Leal-Junior, Paulo de Tarso Camillo de Carvalho, Effects and parameters of the photobiomodulation in experimental models of third-degree burn: systematic review, 2019, 34, 0268-8921, 637, 10.1007/s10103-018-2633-3 | |
46. | Eloá F. Yamada, Franciane Bobinski, Daniel F. Martins, Juliete Palandi, Vanderlei Folmer, Morgana D. da Silva, Photobiomodulation therapy in knee osteoarthritis reduces oxidative stress and inflammatory cytokines in rats, 2020, 13, 1864-063X, 10.1002/jbio.201900204 | |
47. | Cyprien Guermonprez, Lieve Declercq, Géraldine Decaux, Jean‐Alexis Grimaud, Safety and efficacy of a novel home‐use device for light‐potentiated ( LED ) skin treatment , 2020, 13, 1864-063X, 10.1002/jbio.202000230 | |
48. | Kosta Arger, John C. Castel, Dawn Castel, Bernard Choi, Haishan Zeng, 2019, A novel approach to acne treatment utilizing topical printed LED pulsed blue light (Conference Presentation), 9781510623446, 11, 10.1117/12.2509063 | |
49. | Atarodsadat Mostafavinia, Mohammad Bidram, Amirhossein Gomi Avili, Mohammadamin Mahmanzar, Seyed Ali Karimifard, Ensieh Sajadi, Abdollah Amini, Mahsa Hadipour Jahromy, Seyed Kamran Ghoreishi, Sufan Chien, Mohammad Bayat, An improvement in acute wound healing in rats by the synergistic effect of photobiomodulation and arginine, 2019, 35, 2233-7660, 10.1186/s42826-019-0025-x | |
50. | Gaurav K. Keshri, Gaurav Kumar, Manish Sharma, Kiran Bora, Bhuvnesh Kumar, Asheesh Gupta, Photobiomodulation effects of pulsed-NIR laser (810 nm) and LED (808 ± 3 nm) with identical treatment regimen on burn wound healing: A quantitative label-free global proteomic approach, 2021, 6, 26664690, 100024, 10.1016/j.jpap.2021.100024 | |
51. | Marwan El Mobadder, Fadi Farhat, Wassim El Mobadder, Samir Nammour, Photobiomodulation Therapy in the Treatment of Oral Mucositis, Dysphagia, Oral Dryness, Taste Alteration, and Burning Mouth Sensation Due to Cancer Therapy: A Case Series, 2019, 16, 1660-4601, 4505, 10.3390/ijerph16224505 | |
52. | Ann Liebert, Emerging Applications of Photobiomodulation Therapy: The Interaction Between Metabolomics and the Microbiome, 2018, 36, 1557-8550, 515, 10.1089/pho.2018.4527 | |
53. | Mark Cronshaw, Steven Parker, Eugenia Anagnostaki, Valina Mylona, Edward Lynch, Martin Grootveld, Photobiomodulation Dose Parameters in Dentistry: A Systematic Review and Meta-Analysis, 2020, 8, 2304-6767, 114, 10.3390/dj8040114 | |
54. | Sajan George, Michael R. Hamblin, Heidi Abrahamse, Effect of red light and near infrared laser on the generation of reactive oxygen species in primary dermal fibroblasts, 2018, 188, 10111344, 60, 10.1016/j.jphotobiol.2018.09.004 | |
55. | Steven Parker, Eugenia Anagnostaki, Valina Mylona, Mark Cronshaw, Edward Lynch, Martin Grootveld, Systematic Review of Post-Surgical Laser-Assisted Oral Soft Tissue Outcomes Using Surgical Wavelengths Outside the 650–1350 nm Optical Window, 2020, 38, 2578-5478, 591, 10.1089/photob.2020.4847 | |
56. | Nicholas Alexander Wise, 2019, 9780128153055, 489, 10.1016/B978-0-12-815305-5.00036-1 | |
57. | Angela Domínguez Camacho, Sergio Andrés Velásquez, Neftalí Joaquín Benjumea Marulanda, Mauricio Moreno, Photobiomodulation as oedema adjuvant in post-orthognathic surgery patients: A randomized clinical trial, 2020, 18, 17617227, 69, 10.1016/j.ortho.2019.09.004 | |
58. | Titov Vladimir, Osipov Anatoly, Ibragimova Larisa, Petrov Vladimir, Dolgorukova Anna, Oleshkevich Аnna, Hypothetical mechanism of light action on nitric oxide physiological effects, 2020, 0268-8921, 10.1007/s10103-020-03169-x | |
59. | Michèle Pelletier‐Aouizerate, Yvona Zivic, Early cases of acute infectious respiratory syndrome treated with photobiomodulation, diagnosis and intervention: Two case reports, 2021, 2050-0904, 10.1002/ccr3.4058 | |
60. | Francislene de Fatima Cordeiro Petz, Jorge Vinicius Cestari Félix, Hellen Roehrs, Franciele Soares Pott, Janislei Giseli Dorociaki Stocco, Rodrigo Labat Marcos, Marineli Joaquim Meier, Effect of Photobiomodulation on Repairing Pressure Ulcers in Adult and Elderly Patients: A Systematic Review, 2020, 96, 0031-8655, 191, 10.1111/php.13162 | |
61. | Steffen Stein, Michael Schauseil, Andreas Hellak, Heike Korbmacher-Steiner, Andreas Braun, Influence of Photobiomodulation Therapy on Gingivitis Induced by Multi-Bracket Appliances: A Split-Mouth Randomized Controlled Trial, 2018, 36, 1557-8550, 399, 10.1089/pho.2017.4404 | |
62. | Andre D. Paredes, David Benavidez, Jun Cheng, Steve Mangos, Rachana Patil, Michael Donoghue, Enrico Benedetti, Amelia Bartholomew, The Effect of Fluence on Macrophage Kinetics, Oxidative Stress, and Wound Closure Using Real-Time In Vivo Imaging, 2019, 37, 2578-5478, 45, 10.1089/photob.2018.4494 | |
63. | Arun G. Maiya, A. Sampath Kumar, Animesh Hazari, Radhika Jadhav, L Ramachandra, H. Manjunatha Hande, Shenoy K Rajgopal, Shubha G. Maiya, Prabath Kalkura, Laxmikant G. Keni, Photobiomodulation therapy in neuroischaemic diabetic foot ulcers: a novel method of limb salvage, 2018, 27, 0969-0700, 837, 10.12968/jowc.2018.27.12.837 | |
64. | Cesar Augusto Migliorati, 2020, Chapter 4, 978-3-030-29603-2, 37, 10.1007/978-3-030-29604-9_4 | |
65. | Tatiana Dias Schalch, Maria Helena Fernandes, Maria Fernanda Setúbal Destro Rodrigues, Douglas Magno Guimarães, Fabio Daumas Nunes, João Costa Rodrigues, Mônica Pereira Garcia, Raquel Agnelli Mesquita Ferrari, Sandra Kalil Bussadori, Kristianne Porta Santos Fernandes, Photobiomodulation is associated with a decrease in cell viability and migration in oral squamous cell carcinoma, 2019, 34, 0268-8921, 629, 10.1007/s10103-018-2640-4 | |
66. | Hoon Kim, Hong-Bee Kim, Jae-Hwi Seo, Hongbin Kim, Kyong Jin Cho, Effect of 808-nm Laser Photobiomodulation Treatment in Blepharitis Rat Model, 2021, 40, 0277-3740, 358, 10.1097/ICO.0000000000002596 | |
67. | Sandy W. Jere, Nicolette N. Houreld, Heidi Abrahamse, Effect of photobiomodulation on cellular migration and survival in diabetic and hypoxic diabetic wounded fibroblast cells, 2021, 36, 0268-8921, 365, 10.1007/s10103-020-03041-y | |
68. | Iuliia Golovynska, Sergii Golovynskyi, Yurii V. Stepanov, Liudmyla I. Stepanova, Junle Qu, Tymish Y. Ohulchanskyy, Red and near-infrared light evokes Ca2+ influx, endoplasmic reticulum release and membrane depolarization in neurons and cancer cells, 2021, 214, 10111344, 112088, 10.1016/j.jphotobiol.2020.112088 | |
69. | Photobiomodulation Mitigates Cerebrovascular Leakage Induced by the Parkinsonian Neurotoxin MPTP, 2019, 9, 2218-273X, 564, 10.3390/biom9100564 | |
70. | Farzad Salehpour, Javad Mahmoudi, Saeed Sadigh-Eteghad, Paolo Cassano, 2019, 9780128153055, 189, 10.1016/B978-0-12-815305-5.00014-2 | |
71. | Rafał B. Lewandowski, Małgorzata Stępińska, Andrzej Gietka, Monika Dobrzyńska, Mariusz P. Łapiński, Elżbieta A. Trafny, The red-light emitting diode irradiation increases proliferation of human bone marrow mesenchymal stem cells preserving their immunophenotype, 2021, 0955-3002, 1, 10.1080/09553002.2021.1876947 | |
72. | Flaminia Chellini, Alessia Tani, Sandra Zecchi-Orlandini, Marco Giannelli, Chiara Sassoli, In Vitro Evidences of Different Fibroblast Morpho-Functional Responses to Red, Near-Infrared and Violet-Blue Photobiomodulation: Clues for Addressing Wound Healing, 2020, 10, 2076-3417, 7878, 10.3390/app10217878 | |
73. | Susie Suh, Elliot H. Choi, Natasha Atanaskova Mesinkovska, The expression of opsins in the human skin and its implications for photobiomodulation: A Systematic Review, 2020, 36, 0905-4383, 329, 10.1111/phpp.12578 | |
74. | Ali Moradi, Fatemeh Zare, Atarodsadat Mostafavinia, Sobhan Safaju, Amirhossein Shahbazi, Malihe Habibi, Mohammad-Amin Abdollahifar, Seyed Mahmoud Hashemi, Abdollah Amini, Seyed Kamran Ghoreishi, Sufan Chien, Michael R. Hamblin, Reza Kouhkheil, Mohammad Bayat, Photobiomodulation plus Adipose-derived Stem Cells Improve Healing of Ischemic Infected Wounds in Type 2 Diabetic Rats, 2020, 10, 2045-2322, 10.1038/s41598-020-58099-z | |
75. | Anju Yadav, Saurabh Verma, Gaurav K. Keshri, Asheesh Gupta, Role of 904 nm superpulsed laser‐mediated photobiomodulation on nitroxidative stress and redox homeostasis in burn wound healing, 2020, 36, 0905-4383, 208, 10.1111/phpp.12538 | |
76. | Carlo Grandi, Maria Concetta D’Ovidio, Balance between Health Risks and Benefits for Outdoor Workers Exposed to Solar Radiation: An Overview on the Role of Near Infrared Radiation Alone and in Combination with Other Solar Spectral Bands, 2020, 17, 1660-4601, 1357, 10.3390/ijerph17041357 | |
77. | Claudiane Pedro Rodrigues, Jeferson Lucas Jacinto, Mirela Casonato Roveratti, João Pedro Nunes, Francis Lopes Pacagnelli, Rodrigo Antonio Carvalho Andraus, Lúcio Flávio Soares-Caldeira, Alex Silva Ribeiro, Cosme Franklim Buzzachera, Andreo Fernando Aguiar, Effects of Laser Photobiomodulation Therapy at 808 nm on Muscle Performance and Perceived Exertion in Elderly Women, 2020, 36, 0882-7524, 237, 10.1097/TGR.0000000000000288 | |
78. | Xiaoshuang Zuo, Zhuowen Liang, Jiawei Zhang, Shuang Wang, Qiao Zheng, Yangguang Ma, Penghui Li, Tan Ding, Xueyu Hu, Zhe Wang, Photobiomodulation and diffusing optical fiber on spinal cord’s impact on nerve cells from normal spinal cord tissue in piglets, 2021, 0268-8921, 10.1007/s10103-020-03231-8 | |
79. | Elineides Santos Silva, Gabriela Benedito Machado, Adriana Lino-dos-Santos-Franco, Christiane Pavani, Combination of Natural Extracts and Photobiomodulation in Keratinocytes Subjected to UVA Radiation, 2019, 95, 00318655, 644, 10.1111/php.13026 | |
80. | Hye‐Eun Kim, Sayemul Islam, Moonchul Park, Albert Kim, Geelsu Hwang, A Comprehensive Analysis of Near‐Contact Photobiomodulation Therapy in the Host–Bacteria Interaction Model Using 3D‐Printed Modular LED Platform, 2020, 4, 2366-7478, 1900227, 10.1002/adbi.201900227 | |
81. | Laura Marinela Ailioaie, Gerhard Litscher, Curcumin and Photobiomodulation in Chronic Viral Hepatitis and Hepatocellular Carcinoma, 2020, 21, 1422-0067, 7150, 10.3390/ijms21197150 | |
82. | Patricia Gabrielli Vassão, Mayra Cavenague de Souza, Bruna Arcaim Silva, Rheguel Grillo Junqueira, Marcela Regina de Camargo, Victor Zuniga Dourado, Helga Tatiana Tucci, Ana Claudia Renno, Photobiomodulation via a cluster device associated with a physical exercise program in the level of pain and muscle strength in middle-aged and older women with knee osteoarthritis: a randomized placebo-controlled trial, 2020, 35, 0268-8921, 139, 10.1007/s10103-019-02807-3 | |
83. | Shirley Genah, Francesca Cialdai, Valerio Ciccone, Elettra Sereni, Lucia Morbidelli, Monica Monici, Effect of NIR Laser Therapy by MLS-MiS Source on Fibroblast Activation by Inflammatory Cytokines in Relation to Wound Healing, 2021, 9, 2227-9059, 307, 10.3390/biomedicines9030307 | |
84. | Valdison P. Reis, Cristina M.A. Rego, Sulamita S. Setúbal, Maria Naiara M. Tavares, Charles N. Boeno, Alex A. Ferreira e Ferreira, Mauro V. Paloschi, Andreimar M. Soares, Stella R. Zamuner, Juliana P. Zuliani, Effect of light emitting diode photobiomodulation on murine macrophage function after Bothrops envenomation, 2021, 333, 00092797, 109347, 10.1016/j.cbi.2020.109347 | |
85. | Gaurav K. Keshri, Anju Yadav, Saurabh Verma, Bhuvnesh Kumar, Asheesh Gupta, Effects of Pulsed 810 nm Al‐Ga‐As Diode Laser on Wound Healing Under Immunosuppression: A Molecular Insight, 2020, 52, 0196-8092, 424, 10.1002/lsm.23156 | |
86. | E. S. Tuchina, V. V. Tuchin, Photothermal Effect of Infrared (808 nm) Laser Radiation and Gold Nanoparticles in Different Modifications on S. aureus, 2020, 128, 0030-400X, 843, 10.1134/S0030400X20060223 | |
87. | Patricia Gabrielli Vassão, Renata Luri Toma, Hanna Karen Moreira Antunes, Ana Claudia Muniz Renno, Photobiomodulation and physical exercise on strength, balance and functionality of elderly women, 2018, 31, 1980-5918, 10.1590/1980-5918.031.ao09 | |
88. | Lisa A. Miller, Debbie (Gross) Torraca, Luis De Taboada, Retrospective Observational Study and Analysis of Two Different Photobiomodulation Therapy Protocols Combined with Rehabilitation Therapy as Therapeutic Interventions for Canine Degenerative Myelopathy, 2020, 38, 2578-5478, 195, 10.1089/photob.2019.4723 | |
89. | Patrick Benson, Ji Yeon Kim, Carlos Riveros, Aaron Camp, Daniel M. Johnstone, Elucidating the time course of the transcriptomic response to photobiomodulation through gene co-expression analysis, 2020, 208, 10111344, 111916, 10.1016/j.jphotobiol.2020.111916 | |
90. | Ewa Jówko, Maciej Płaszewski, Maciej Cieśliński, Tomasz Sacewicz, Igor Cieśliński, Marta Jarocka, The effect of low level laser irradiation on oxidative stress, muscle damage and function following neuromuscular electrical stimulation. A double blind, randomised, crossover trial, 2019, 11, 2052-1847, 10.1186/s13102-019-0147-3 | |
91. | Narda G. Robinson, Beyond the Laboratory, Into the Clinic: What Dogs with Disk Disease Have Taught Us About Photobiomodulation for Spinal Cord Injury, 2017, 35, 1557-8550, 589, 10.1089/pho.2017.4348 | |
92. | Anand Marya, Adith Venugopal, Nafij Jamayet, The Use of Technology in the Management of Orthodontic Treatment-Related Pain, 2021, 2021, 1918-1523, 1, 10.1155/2021/5512031 | |
93. | Mateus Rossato, Rodolfo A. Dellagrana, Raphael L. Sakugawa, Caetano D. Lazzari, Bruno M. Baroni, Fernando Diefenthaeler, Time Response of Photobiomodulation Therapy on Muscular Fatigue in Humans, 2018, 32, 1064-8011, 3285, 10.1519/JSC.0000000000002339 | |
94. | Mariia Lunova, Barbora Smolková, Mariia Uzhytchak, Klára Žofie Janoušková, Milan Jirsa, Daria Egorova, Andrei Kulikov, Šárka Kubinová, Alexandr Dejneka, Oleg Lunov, Light-induced modulation of the mitochondrial respiratory chain activity: possibilities and limitations, 2020, 77, 1420-682X, 2815, 10.1007/s00018-019-03321-z | |
95. | Priscila Thaís Rodrigues de Abreu, José Alcides Almeida de Arruda, Ricardo Alves Mesquita, Lucas Guimarães Abreu, Ivana Márcia Alves Diniz, Tarcília Aparecida Silva, Photobiomodulation effects on keratinocytes cultured in vitro: a critical review, 2019, 34, 0268-8921, 1725, 10.1007/s10103-019-02813-5 | |
96. | Farzad Salehpour, Javad Mahmoudi, Farzin Kamari, Saeed Sadigh-Eteghad, Seyed Hossein Rasta, Michael R Hamblin, Brain Photobiomodulation Therapy: a Narrative Review, 2018, 55, 0893-7648, 6601, 10.1007/s12035-017-0852-4 | |
97. | Vahid Mansouri, Mohammadreza Razzaghi, Mohammad Rostami-Nejad, Majid Rezaei-Tavirani, Mohammad Hossein Heidari, Saeed Safari, Babak Arjmand, Mostafa Rezaei-Tavirani, Alireza Zali, Mostafa Hamdieh, Neuroprotective Properties of Photobiomodulation in Retinal Regeneration in Rats: Perspectives From Interaction Levels, 2020, 11, 2008-9783, 280, 10.34172/jlms.2020.47 | |
98. | Elisabetta Merigo, Jean-Paul Rocca, Antonio L.B. Pinheiro, Carlo Fornaini, Photobiomodulation Therapy in Oral Medicine: A Guide for the Practitioner with Focus on New Possible Protocols, 2019, 37, 2578-5478, 669, 10.1089/photob.2019.4624 | |
99. | Sandy W. Jere, Nicolette N. Houreld, Heidi Abrahamse, Role of the PI3K/AKT (mTOR and GSK3β) signalling pathway and photobiomodulation in diabetic wound healing, 2019, 50, 13596101, 52, 10.1016/j.cytogfr.2019.03.001 | |
100. | A.N. Otterço, A.L. Andrade, P. Brassolatti, K.N.Z. Pinto, H.S.S. Araújo, N.A. Parizotto, Photobiomodulation mechanisms in the kinetics of the wound healing process in rats, 2018, 183, 10111344, 22, 10.1016/j.jphotobiol.2018.04.010 | |
101. | Natalia Arias, Juan Díaz González, Alberto Martín Pernía, Jorge L. Arias, 2019, 9780128153055, 253, 10.1016/B978-0-12-815305-5.00020-8 | |
102. | Gopal Nambi, Does low level laser therapy has effects on inflammatory biomarkers IL-1β, IL-6, TNF-α, and MMP-13 in osteoarthritis of rat models—a systemic review and meta-analysis, 2021, 36, 0268-8921, 475, 10.1007/s10103-020-03124-w | |
103. | Bruna Hoffmann de Oliveira, Verônica Vargas Horewicz, Rafaela Hardt da Silva, Daiana Cristina Salm, Afonso S.I. Salgado, Francisco José Cidral-Filho, Franciane Bobinski, Anna Paula Piovezan, Daniel F. Martins, ET-B receptors involvement in peripheral opioid analgesia induced by light-emitting diode photobiomodulation in male and female mice, 2021, 214, 10111344, 112104, 10.1016/j.jphotobiol.2020.112104 | |
104. | Fabrízio dos Santos Cardoso, Cristiane de Souza Oliveira Tavares, Bruno Henrique Silva Araujo, Fernanda Mansur, Rodrigo Álvaro Brandão Lopes-Martins, Sérgio Gomes da Silva, Improved Spatial Memory And Neuroinflammatory Profile Changes in Aged Rats Submitted to Photobiomodulation Therapy, 2021, 0272-4340, 10.1007/s10571-021-01069-4 | |
105. | Gabriela Silva, Cleber Ferraresi, Rodrigo T. Almeida, Mariana L. Motta, Thiago Paixão, Vinicius O. Ottone, Ivana A. Fonseca, Murilo X. Oliveira, Etel Rocha‐Vieira, Marco F. Dias‐Peixoto, Elizabethe A. Esteves, Cândido C. Coimbra, Fabiano T. Amorim, Flávio de Castro Magalhães, Insulin resistance is improved in high‐fat fed mice by photobiomodulation therapy at 630 nm, 2020, 13, 1864-063X, 10.1002/jbio.201960140 | |
106. | Paula I. Martin, Linda Chao, Maxine H. Krengel, Michael D. Ho, Megan Yee, Robert Lew, Jeffrey Knight, Michael R. Hamblin, Margaret A. Naeser, Transcranial Photobiomodulation to Improve Cognition in Gulf War Illness, 2021, 11, 1664-2295, 10.3389/fneur.2020.574386 | |
107. | Auriléia Aparecida de Brito, Elaine Cristina da Silveira, Nicole Cristine Rigonato-Oliveira, Stephanie Souza Soares, Maysa Alves Rodrigues Brandao-Rangel, Clariana Rodrigues Soares, Tawany Gonçalves Santos, Cintia Estefano Alves, Karine Zanella Herculano, Rodolfo Paula Vieira, Adriana Lino-dos-Santos-Franco, Regiane Albertini, Flavio Aimbire, Ana Paula de Oliveira, Low-level laser therapy attenuates lung inflammation and airway remodeling in a murine model of idiopathic pulmonary fibrosis: Relevance to cytokines secretion from lung structural cells, 2020, 203, 10111344, 111731, 10.1016/j.jphotobiol.2019.111731 | |
108. | Weyland Cheng, Manye Yao, Keming Sun, Weili Li, Progress in Photobiomodulation for Bone Fractures: A Narrative Review, 2020, 38, 2578-5478, 260, 10.1089/photob.2019.4732 | |
109. | Qiao Zheng, Jiawei Zhang, Xiaoshuang Zuo, Jiakai Sun, Zhuowen Liang, Xueyu Hu, Zhe Wang, Kun Li, Jiwei Song, Tan Ding, Xuefeng Shen, Yangguang Ma, Penghui Li, Photobiomodulation Promotes Neuronal Axon Regeneration After Oxidative Stress and Induces a Change in Polarization from M1 to M2 in Macrophages via Stimulation of CCL2 in Neurons: Relevance to Spinal Cord Injury, 2021, 0895-8696, 10.1007/s12031-020-01756-9 | |
110. | Min Ho Hwang, Jae Won Lee, Hyeong-Guk Son, Joohan Kim, Hyuk Choi, Effects of photobiomodulation on annulus fibrosus cells derived from degenerative disc disease patients exposed to microvascular endothelial cells conditioned medium, 2020, 10, 2045-2322, 10.1038/s41598-020-66689-0 | |
111. | Ikonija Koceva, Bettina Rümmelein, Peter Arne Gerber, Deirdre Edge, Michael Canova Engelbrecht Nielsen, Fluorescent light energy: A new therapeutic approach to effectively treating acne conglobata and hidradenitis suppurativa, 2019, 7, 2050-0904, 1769, 10.1002/ccr3.2334 | |
112. | Antonio Russo, Maiken Mellergaard, Giovanni Pellacani, Steven Nisticò, Michael Canova Engelbrecht Nielsen, Fluorescent light energy combined with systemic isotretinoin: A 52‐week follow‐up evaluating efficacy and safety in treatment of moderate‐severe acne, 2021, 2050-0904, 10.1002/ccr3.3944 | |
113. | Aline Barbosa Macedo, Daniela Sayuri Mizobuti, Tulio de Almeida Hermes, Rafael Dias Mâncio, Adriana Pertille, Larissa Akemi Kido, Valéria Helena Alves Cagnon, Elaine Minatel, Photobiomodulation Therapy for Attenuating the Dystrophic Phenotype of MdxMice, 2020, 96, 0031-8655, 200, 10.1111/php.13179 | |
114. | Justin H. Rigby, Austin M. Hagan, A Novel Blue–Red Photobiomodulation Therapy Patch Effects on a Repetitive Elbow-Flexion Fatigue Task, 2020, 29, 1056-6716, 271, 10.1123/jsr.2018-0339 | |
115. | Sungkyoo Lim, Suk-Jun Lee, Eal-Whan Park, Michael R. Hamblin, James D. Carroll, Praveen Arany, 2019, Fat loss by red and near infrared LED phototherapy, 9781510623644, 11, 10.1117/12.2506800 | |
116. | Zeqing Chen, Wenqi Li, Xiaojian Hu, Muqing Liu, Irradiance plays a significant role in photobiomodulation of B16F10 melanoma cells by increasing reactive oxygen species and inhibiting mitochondrial function, 2020, 11, 2156-7085, 27, 10.1364/BOE.11.000027 | |
117. | Rosane de Fatima Zanirato Lizarelli, Vanderlei Salvador Bagnato, 2020, Chapter 6, 978-3-030-29603-2, 59, 10.1007/978-3-030-29604-9_6 | |
118. | A. N. Otterço, P. Brassolatti, A. L. M. Andrade, L. R. S. Avó, P. S. Bossini, N. A. E. Parizotto, Effect of photobiomodulation (670 nm) associated with vitamin A on the inflammatory phase of wound healing, 2018, 33, 0268-8921, 1867, 10.1007/s10103-018-2535-4 | |
119. | Andrew McColloch, Caleb Liebman, Hanli Liu, Michael Cho, Alterted Adipogenesis of Human Mesenchymal Stem Cells by Photobiomodulation Using 1064 nm Laser Light, 2021, 53, 0196-8092, 263, 10.1002/lsm.23278 | |
120. | Angela Maria Paiva Magri, Kelly Rossetti Fernandes, Hueliton Wilian Kido, Gabriela Sodano Fernandes, Stephanie de Souza Fermino, Paulo Roberto Gabbai-Armelin, Franscisco José Correa Braga, Cintia Pereira de Góes, José Lucas dos Santos Prado, Renata Neves Granito, Ana Claudia Muniz Rennó, Photobiomodulation guided healing in a sub-critical bone defect in calvarias of rats, 2019, 28, 0898-5901, 171, 10.5978/islsm.28_19-OR-13 | |
121. | Hongjun Wu, Ziyang Zang, Zhenhua Pan, Jia Shi, Hongli Chen, Zhibo Han, Jianquan Yao, Combined Effects of Low Level Laser Therapy and Inducers on the Neural Differentiation of Mesenchymal Stem Cells, 2021, 9, 2169-3536, 28946, 10.1109/ACCESS.2021.3052942 | |
122. | Tomasz Walski, Krystyna Dąbrowska, Anna Drohomirecka, Natalia Jędruchniewicz, Natalia Trochanowska-Pauk, Wojciech Witkiewicz, Małgorzata Komorowska, The effect of red-to-near-infrared (R/NIR) irradiation on inflammatory processes, 2019, 95, 0955-3002, 1326, 10.1080/09553002.2019.1625464 | |
123. | Jiayu Wang, Jianfei Dong, Optical Waveguides and Integrated Optical Devices for Medical Diagnosis, Health Monitoring and Light Therapies, 2020, 20, 1424-8220, 3981, 10.3390/s20143981 | |
124. |
Chaiyarerk Homsirikamol, Saroj Suvanasuthi, Kwanchanok Viravaidya-Pasuwat,
Inclusion of IR-820 into Soybean-Phosphatides-Based Nanoparticles for Near-Infrared-Triggered Release and Endolysosomal Escape in HaCaT Keratinocytes at Insignificant Cytotoxic Level , 2020, Volume 15, 1178-2013, 8717, 10.2147/IJN.S267119 |
|
125. | L. Cristiano, Use of infrared-based devices in aesthetic medicine and for beauty and wellness treatments, 2019, 102, 13504495, 102991, 10.1016/j.infrared.2019.102991 | |
126. | Iuliia Golovynska, Sergii Golovynskyi, Yurii V. Stepanov, Liudmyla V. Garmanchuk, Ludmila I. Stepanova, Junle Qu, Tymish Y. Ohulchanskyy, Red and near‐infrared light induces intracellular Ca 2+ flux via the activation of glutamate N ‐methyl‐D‐aspartate receptors , 2019, 234, 0021-9541, 15989, 10.1002/jcp.28257 | |
127. | Mohammad Bagheri, Atarodsadat Mostafavinia, Mohammad-Amin Abdollahifar, Abdollah Amini, Seyed Kamran Ghoreishi, Sufan Chien, Michael R. Hamblin, Sahar Bayat, Mohammad Bayat, Combined effects of metformin and photobiomodulation improve the proliferation phase of wound healing in type 2 diabetic rats, 2020, 123, 07533322, 109776, 10.1016/j.biopha.2019.109776 | |
128. | Sungkyoo Lim, Eal-Whan Park, Michael R. Hamblin, James D. Carroll, Praveen Arany, 2018, Spot fat reduction by red and near infrared LED phototherapy, 9781510614390, 10, 10.1117/12.2289386 | |
129. | Tamiris Silva, Yara Dadalti Fragoso, Maria Fernanda Setúbal Destro Rodrigues, Andréa Oliver Gomes, Fernanda Cordeiro da Silva, Lucas Andreo, Ariane Viana, Daniela de Fátima Teixeira da Silva, Maria Cristina Chavantes, Anna Carolina Ratto Tempestini Horliana, Kátia De Angelis, Alessandro Melo Deana, Luciana Prats Branco, Kristianne Porta Santos Fernandes, Lara Jansiski Motta, Raquel Agnelli Mesquita-Ferrari, Sandra Kalil Bussadori, Tim Friede, Effects of photobiomodulation on interleukin-10 and nitrites in individuals with relapsing-remitting multiple sclerosis – Randomized clinical trial, 2020, 15, 1932-6203, e0230551, 10.1371/journal.pone.0230551 | |
130. | Houssein Ahmadi, Abdollah Amini, Fatemeh Fadaei Fathabady, Atarodsadat Mostafavinia, Fatemeh Zare, Roohollah Ebrahimpour-malekshah, Mustafa Neshat Ghalibaf, Matin Abrisham, Fatemehalsadat Rezaei, Richard Albright, Seyed Kamran Ghoreishi, Sufan Chien, Mohammad Bayat, Transplantation of photobiomodulation-preconditioned diabetic stem cells accelerates ischemic wound healing in diabetic rats, 2020, 11, 1757-6512, 10.1186/s13287-020-01967-2 | |
131. | Tainá Caroline Santos, Kaline Brito Sousa, Lucas Andreo, Andreia Martinelli, Maria Fernanda Setubal Destro Rodrigues, Sandra Kalil Bussadori, Kristianne Porta Santos Fernandes, Raquel Agnelli Mesquita‐Ferrari, Effect of Photobiomodulation on C2C12 Myoblasts Cultivated in M1 Macrophage‐conditioned Media, 2020, 96, 0031-8655, 906, 10.1111/php.13215 | |
132. | Frederico Carlos Jana Neto, Ana Luiza Cabrera Martimbianco, Raíssa Piagentini de Andrade, Sandra Kalil Bussadori, Raquel Agnelli Mesquita-Ferrari, Kristianne Porta Santos Fernandes, Effects of photobiomodulation in the treatment of fractures: a systematic review and meta-analysis of randomized clinical trials, 2020, 35, 0268-8921, 513, 10.1007/s10103-019-02779-4 | |
133. | Charles Mignon, Natallia E. Uzunbajakava, Irene Castellano-Pellicena, Natalia V. Botchkareva, Desmond J. Tobin, Differential response of human dermal fibroblast subpopulations to visible and near-infrared light: Potential of photobiomodulation for addressing cutaneous conditions, 2018, 50, 01968092, 859, 10.1002/lsm.22823 | |
134. | Yujin Ohsugi, Hiromi Niimi, Tsuyoshi Shimohira, Masahiro Hatasa, Sayaka Katagiri, Akira Aoki, Takanori Iwata, In Vitro Cytological Responses against Laser Photobiomodulation for Periodontal Regeneration, 2020, 21, 1422-0067, 9002, 10.3390/ijms21239002 | |
135. | Raquel Cantero-Téllez, Jorge Hugo Villafañe, Kirstin Valdes, Santiago García-Orza, Mark D Bishop, Ivan Medina-Porqueres, Effects of High-Intensity Laser Therapy on Pain Sensitivity and Motor Performance in Patients with Thumb Carpometacarpal Joint Osteoarthritis: A Randomized Controlled Trial, 2020, 21, 1526-2375, 2357, 10.1093/pm/pnz297 | |
136. | Songyun Wang, Qinyu Luo, Hui Chen, Jingyu Huang, Xuemeng Li, Lin Wu, Binxun Li, Zhen Wang, Dongdong Zhao, Hong Jiang, Light Emitting Diode Therapy Protects against Myocardial Ischemia/Reperfusion Injury through Mitigating Neuroinflammation, 2020, 2020, 1942-0900, 1, 10.1155/2020/9343160 | |
137. | Taisa Nogueira Pansani, Fernanda Gonçalves Basso, Diana Gabriela Soares, Ana Paula da Silveira Turrioni, Josimeri Hebling, Carlos Alberto de Souza Costa, Photobiomodulation in the Metabolism of Lipopolysaccharides-exposed Epithelial Cells and Gingival Fibroblasts, 2018, 94, 00318655, 598, 10.1111/php.12877 | |
138. | Farzad Salehpour, Fereshteh Farajdokht, Paolo Cassano, Saeed Sadigh-Eteghad, Marjan Erfani, Michael R. Hamblin, Maryam Moghadam Salimi, Pouran Karimi, Seyed Hossein Rasta, Javad Mahmoudi, Near-infrared photobiomodulation combined with coenzyme Q10 for depression in a mouse model of restraint stress: reduction in oxidative stress, neuroinflammation, and apoptosis, 2019, 144, 03619230, 213, 10.1016/j.brainresbull.2018.10.010 | |
139. | Steven Parker, Mark Cronshaw, Eugenia Anagnostaki, Valina Mylona, Edward Lynch, Martin Grootveld, Current Concepts of Laser–Oral Tissue Interaction, 2020, 8, 2304-6767, 61, 10.3390/dj8030061 | |
140. | Steven Shanks, Gerry Leisman, 2018, Chapter 188, 978-3-319-95707-4, 41, 10.1007/5584_2018_188 | |
141. | Agnieszka Gerkowicz, Joanna Bartosińska, Katarzyna Wolska-Gawron, Małgorzata Michalska-Jakubus, Mirosław Kwaśny, Dorota Krasowska, Application of superluminescent diodes (sLED) in the treatment of scarring alopecia – A pilot study, 2019, 28, 15721000, 195, 10.1016/j.pdpdt.2019.09.006 | |
142. | Laura Marinela Ailioaie, Gerhard Litscher, Molecular and Cellular Mechanisms of Arthritis in Children and Adults: New Perspectives on Applied Photobiomodulation, 2020, 21, 1422-0067, 6565, 10.3390/ijms21186565 | |
143. | Alba Gutiérrez-Menéndez, Sandra Cid-Duarte, María Banqueri, Juan A. Martínez, Marta Méndez, Jorge L. Arias, Photobiomodulation effects on active brain networks during a spatial memory task, 2021, 230, 00319384, 113291, 10.1016/j.physbeh.2020.113291 | |
144. | Andrea Marchegiani, Adolfo M. Tambella, Alessandro Fruganti, Andrea Spaterna, Matteo Cerquetella, Susan Paterson, Management of canine perianal fistula with fluorescence light energy: preliminary findings, 2020, 31, 0959-4493, 460, 10.1111/vde.12890 | |
145. | Flávio Klinpovous Kerppers, Kesia Maria Mangoni Gonçalves dos Santos, Maria Elvira Ribeiro Cordeiro, Mário César da Silva Pereira, Danilo Barbosa, André Alexandre Pezzini, Luiza Ferreira Cunha, Maiara Fonseca, Ketlin Bragnholo, Afonso Shiguemi Inoue Salgado, Ivo Ilvan Kerppers, Study of transcranial photobiomodulation at 945-nm wavelength: anxiety and depression, 2020, 35, 0268-8921, 1945, 10.1007/s10103-020-02983-7 | |
146. | Kristýna Kárová, Lucia Machova Urdzíková, Nataliya Romanyuk, Barbora Svobodová, Kristýna Kekulová, Zuzana Kočí, Pavla Jendelová, Šárka Kubinová, 2020, 9780128180846, 291, 10.1016/B978-0-12-818084-6.00008-8 | |
147. | Ruxandra Elena Luca, Alessandra Giuliani, Adrian Mănescu, Rodica Heredea, Bogdan Hoinoiu, George Dumitru Constantin, Virgil-Florin Duma, Carmen Darinca Todea, Osteogenic Potential of Bovine Bone Graft in Combination with Laser Photobiomodulation: An Ex Vivo Demonstrative Study in Wistar Rats by Cross-Linked Studies Based on Synchrotron Microtomography and Histology, 2020, 21, 1422-0067, 778, 10.3390/ijms21030778 | |
148. | Reza Zomorrodi, Genane Loheswaran, Abhiram Pushparaj, Lew Lim, Pulsed Near Infrared Transcranial and Intranasal Photobiomodulation Significantly Modulates Neural Oscillations: a pilot exploratory study, 2019, 9, 2045-2322, 10.1038/s41598-019-42693-x | |
149. | Thais Ferreira Gomes, Matheus Masalskiene Pedrosa, Ana Claudia Laforga de Toledo, Veridiana Wanshi Arnoni, Mirian dos Santos Monteiro, Davi Cury Piai, Silvia Helena Zacarias Sylvestre, Bruno Ferreira, Bactericide effect of methylene blue associated with low-level laser therapy in Escherichia coli bacteria isolated from pressure ulcers, 2018, 33, 0268-8921, 1723, 10.1007/s10103-018-2528-3 | |
150. | Vinh Van Tran, Minhe Chae, Ju-Young Moon, Young-Chul Lee, Light emitting diodes technology-based photobiomodulation therapy (PBMT) for dermatology and aesthetics: Recent applications, challenges, and perspectives, 2021, 135, 00303992, 106698, 10.1016/j.optlastec.2020.106698 | |
151. | Eiko Nakayama, Toshihiro Kushibiki, Yoshine Mayumi, Masato Tsuchiya, Ryuichi Azuma, Tomoharu Kiyosawa, Miya Ishihara, Photobiomodulation Therapy in Plastic Surgery and Dermatology, 2021, 41, 0288-6200, 370, 10.2530/jslsm.jslsm-41_0035 | |
152. | Denis J. Gendron, Michael R. Hamblin, Applications of Photobiomodulation Therapy to Musculoskeletal Disorders and Osteoarthritis with Particular Relevance to Canada, 2019, 37, 2578-5478, 408, 10.1089/photob.2018.4597 | |
153. | Jolien Robijns, Sandrine Censabella, Stefan Claes, Luc Pannekoeke, Lore Bussé, Dora Colson, Iris Kaminski, Joy Lodewijckx, Paul Bulens, Annelies Maes, Leen Noé, Marc Brosens, An Timmermans, Ivo Lambrichts, Veerle Somers, Jeroen Mebis, Biophysical skin measurements to evaluate the effectiveness of photobiomodulation therapy in the prevention of acute radiation dermatitis in breast cancer patients, 2019, 27, 0941-4355, 1245, 10.1007/s00520-018-4487-4 | |
154. | Ann Liebert, Brian Bicknell, Daniel M. Johnstone, Luke C. Gordon, Hosen Kiat, Michael R. Hamblin, “Photobiomics”: Can Light, Including Photobiomodulation, Alter the Microbiome?, 2019, 37, 2578-5478, 681, 10.1089/photob.2019.4628 | |
155. | Patricia Gabrielli Vassão, Bruna Arcaim Silva, Mayra Cavenague de Souza, Julia Risso Parisi, Marcela Regina de Camargo, Ana Claudia Muniz Renno, Level of pain, muscle strength and posture: effects of PBM on an exercise program in women with knee osteoarthritis – a randomized controlled trial, 2020, 35, 0268-8921, 1967, 10.1007/s10103-020-02989-1 | |
156. | Jolien Robijns, Sandrine Censabella, Stefan Claes, Luc Pannekoeke, Lore Bussé, Dora Colson, Iris Kaminski, Paul Bulens, Annelies Maes, Leen Noé, Marc Brosens, An Timmermans, Ivo Lambrichts, Veerle Somers, Jeroen Mebis, Prevention of acute radiodermatitis by photobiomodulation: A randomized, placebo-controlled trial in breast cancer patients (TRANSDERMIS trial), 2018, 50, 01968092, 763, 10.1002/lsm.22804 | |
157. | Margaret A. Naeser, Michael D. Ho, Paula I. Martin, Michael R. Hamblin, Bang-Bon Koo, Increased Functional Connectivity Within Intrinsic Neural Networks in Chronic Stroke Following Treatment with Red/Near-Infrared Transcranial Photobiomodulation: Case Series with Improved Naming in Aphasia, 2020, 38, 2578-5478, 115, 10.1089/photob.2019.4630 | |
158. | Mahasweta Bhattacharya, Anirban Dutta, Computational Modeling of the Photon Transport, Tissue Heating, and Cytochrome C Oxidase Absorption during Transcranial Near-Infrared Stimulation, 2019, 9, 2076-3425, 179, 10.3390/brainsci9080179 | |
159. | Reza Fekrazad, Photobiomodulation and Antiviral Photodynamic Therapy as a Possible Novel Approach in COVID-19 Management, 2020, 38, 2578-5478, 255, 10.1089/photob.2020.4868 | |
160. | Alexander N. Yakunin, Sergey V. Zarkov, Yuri A. Avetisyan, Garif G. Akchurin, Georgy G. Akchurin, Elena S. Tuchina, Valery V. Tuchin, Valery V. Tuchin, Elina A. Genina, 2019, Modeling of hyperthermia induced by functionalized gold nanorods bound to Staphylococcus aureus under NIR laser radiation, 9781510628205, 107, 10.1117/12.2531440 | |
161. | Massimo Petruzzi, Gianna Maria Nardi, Fabio Cocco, Fedora della Vella, Roberta Grassi, Felice Roberto Grassi, Polarized Polychromatic Noncoherent Light (Bioptron Light) as Adjunctive Treatment in Chronic Oral Mucosal Pain: A Pilot Study, 2019, 37, 2578-5478, 227, 10.1089/photob.2018.4576 | |
162. | Atarodsadat Mostafavinia, Abdollah Amini, Ensieh Sajadi, Houssein Ahmadi, Fatemehalsadat Rezaei, Seyed Kamran Ghoreishi, Sufan Chien, Mohammad Bayat, Photobiomodulation therapy was more effective than photobiomodulation plus arginine on accelerating wound healing in an animal model of delayed healing wound, 2021, 0268-8921, 10.1007/s10103-021-03271-8 | |
163. | Bárbara Argibay, Francisco Campos, María Perez-Mato, Alba Vieites-Prado, Clara Correa-Paz, Esteban López-Arias, Andrés Da Silva-Candal, Vicente Moreno, Carlos Montero, Tomás Sobrino, José Castillo, Ramón Iglesias-Rey, Light-Emitting Diode Photobiomodulation After Cerebral Ischemia, 2019, 10, 1664-2295, 10.3389/fneur.2019.00911 | |
164. | Igor Rafael Correia Rocha, Edward Perez-Reyes, Marucia Chacur, Effect of photobiomodulation on mitochondrial dynamics in peripheral nervous system in streptozotocin-induced type 1 diabetes in rats, 2021, 20, 1474-905X, 293, 10.1007/s43630-021-00018-w | |
165. | Yueying Lu, Jianlou Yang, Chen Dong, Yuming Fu, Hong Liu, Gut microbiome-mediated changes in bone metabolism upon infrared light exposure in rats, 2021, 217, 10111344, 112156, 10.1016/j.jphotobiol.2021.112156 | |
166. | Luodan Yang, Hannah Youngblood, Chongyun Wu, Quanguang Zhang, Mitochondria as a target for neuroprotection: role of methylene blue and photobiomodulation, 2020, 9, 2047-9158, 10.1186/s40035-020-00197-z | |
167. | Binglin Shen, Shiqi Wang, Ganapathi Bharathi, Yanping Li, Fangrui Lin, Rui Hu, Liwei Liu, Junle Qu, Rapid and Targeted Photoactivation of Ca2+ Channels Mediated by Squaraine To Regulate Intracellular and Intercellular Signaling Processes, 2020, 92, 0003-2700, 8497, 10.1021/acs.analchem.0c01243 | |
168. | Saeed Vafaei-Nezhad, Mahnaz Pour Hassan, Mohsen Noroozian, Abbas Aliaghaei, Atefeh Shirazi Tehrani, Hojjat Allah Abbaszadeh, Shahrokh Khoshsirat, A Review of Low-Level Laser Therapy for Spinal Cord Injury: Challenges And Safety, 2020, 11, 2008-9783, 363, 10.34172/jlms.2020.59 | |
169. | Hannah Serrage, Vladimir Heiskanen, William M. Palin, Paul R. Cooper, Michael R. Milward, Mohammed Hadis, Michael R. Hamblin, Under the spotlight: mechanisms of photobiomodulation concentrating on blue and green light, 2019, 18, 1474-905X, 1877, 10.1039/C9PP00089E | |
170. | Mayara Santos de Castro, Marta Miyazawa, Ester Siqueira Caixeta Nogueira, Jorge Kleber Chavasco, Gustavo Andrade Brancaglion, Cláudio Daniel Cerdeira, Denismar Alves Nogueira, Marisa Ionta, João Adolfo Costa Hanemann, Maísa Ribeiro Pereira Lima Brigagão, Felipe Fornias Sperandio, Photobiomodulation enhances the Th1 immune response of human monocytes, 2020, 0268-8921, 10.1007/s10103-020-03179-9 | |
171. | Aude Nicolas, Kevin P. Kenna, Alan E. Renton, Nicola Ticozzi, Faraz Faghri, Ruth Chia, Janice A. Dominov, Brendan J. Kenna, Mike A. Nalls, Pamela Keagle, Alberto M. Rivera, Wouter van Rheenen, Natalie A. Murphy, Joke J.F.A. van Vugt, Joshua T. Geiger, Rick A. Van der Spek, Hannah A. Pliner, Bradley N. Smith, Giuseppe Marangi, Simon D. Topp, Yevgeniya Abramzon, Athina Soragia Gkazi, John D. Eicher, Aoife Kenna, ITALSGEN Consortium, Gabriele Mora, Andrea Calvo, Letizia Mazzini, Nilo Riva, Jessica Mandrioli, Claudia Caponnetto, Stefania Battistini, Paolo Volanti, Vincenzo La Bella, Francesca L. Conforti, Giuseppe Borghero, Sonia Messina, Isabella L. Simone, Francesca Trojsi, Fabrizio Salvi, Francesco O. Logullo, Sandra D’Alfonso, Lucia Corrado, Margherita Capasso, Luigi Ferrucci, Genomic Translation for ALS Care (GTAC) Consortium, Cristiane de Araujo Martins Moreno, Sitharthan Kamalakaran, David B. Goldstein, The ALS Sequencing Consortium, Aaron D. Gitler, Tim Harris, Richard M. Myers, NYGC ALS Consortium, Hemali Phatnani, Rajeeva Lochan Musunuri, Uday Shankar Evani, Avinash Abhyankar, Michael Charles Zody, Answer ALS Foundation, Julia Kaye, Steven Finkbeiner, Stacia Wyman, Alex Lenail, Leandro Lima, Ernest Fraenkel, Clive N. Svendsen, Leslie M. Thompson, Jennifer E. Van Eyk, James D. Berry, Timothy M. Miller, Stephen J. Kolb, Merit Cudkowicz, Emily Baxi, Clinical Research in ALS and Relate Development (CReATe) Consortium, Michael Benatar, J. Paul Taylor, Evadnie Rampersaud, Gang Wu, Joanne Wuu, SLAGEN Consortium, Giuseppe Lauria, Federico Verde, Isabella Fogh, Cinzia Tiloca, Giacomo P. Comi, Gianni Sorarù, Cristina Cereda, French ALS Consortium, Philippe Corcia, Hannu Laaksovirta, Liisa Myllykangas, Lilja Jansson, Miko Valori, John Ealing, Hesham Hamdallah, Sara Rollinson, Stuart Pickering-Brown, Richard W. Orrell, Katie C. Sidle, Andrea Malaspina, John Hardy, Andrew B. Singleton, Janel O. Johnson, Sampath Arepalli, Peter C. Sapp, Diane McKenna-Yasek, Meraida Polak, Seneshaw Asress, Safa Al-Sarraj, Andrew King, Claire Troakes, Caroline Vance, Jacqueline de Belleroche, Frank Baas, Anneloor LMA ten Asbroek, José Luis Muñoz-Blanco, Dena G. Hernandez, Jinhui Ding, J. Raphael Gibbs, Sonja W. Scholz, Mary Kay Floeter, Roy H. Campbell, Francesco Landi, Stefan M. Pulst, John M. Ravits, Daniel J. L. MacGowan, Janine Kirby, Erik Pioro, Roger Pamphlett, James Broach, Glenn Gerhard, Travis L. Dunckley, Christopher B. Brady, Neil W. Kowall, Juan C. Troncoso, Isabelle Le Ber, Kevin Mouzat, Serge Lumbroso, Terry D. Heiman-Patterson, Freya Kamel, Ludo Van Den Bosch, Robert H. Baloh, Tim M. Strom, Thomas Meitinger, Aleksey Shatunov, Kristel R. Van Eijk, Mamede de Carvalho, Maarten Kooyman, Bas Middelkoop, Matthieu Moisse, Russell L. McLaughlin, Michael A. Van Es, Markus Weber, Kevin B. Boylan, Marka Van Blitterswijk, Rosa Rademakers, Karen E. Morrison, A. Nazli Basak, Jesús S. Mora, Vivian E. Drory, Pamela J. Shaw, Martin R. Turner, Kevin Talbot, Orla Hardiman, Kelly L. Williams, Jennifer A. Fifita, Garth A. Nicholson, Ian P. Blair, Guy A. Rouleau, Jesús Esteban-Pérez, Alberto García-Redondo, Ammar Al-Chalabi, Ekaterina Rogaeva, Lorne Zinman, Lyle Ostrow, Nicholas J. Maragakis, Jeffrey D. Rothstein, Zachary Simmons, Johnathan Cooper-Knock, Alexis Brice, Stephen A. Goutman, Eva L. Feldman, Summer B. Gibson, Franco Taroni, Antonia Ratti, Cinzia Gellera, Philip Van Damme, Wim Robberecht, Pietro Fratta, Mario Sabatelli, Christian Lunetta, Albert C. Ludolph, Peter M. Andersen, Jochen H. Weishaupt, William Camu, John Q. Trojanowski, Vivianna M. Van Deerlin, Robert H. Brown, Leonard H. van den Berg, Jan H. Veldink, Matthew B. Harms, Jonathan D. Glass, David J. Stone, Pentti Tienari, Vincenzo Silani, Adriano Chiò, Christopher E. Shaw, Bryan J. Traynor, John E. Landers, Genome-Wide Analyses Identify KIF5A as a Novel ALS Gene, 2018, 1556-5068, 10.2139/ssrn.3155776 | |
172. | Noelia Galiano‐Castillo, Lizhou Liu, Mario Lozano‐Lozano, Steve Tumilty, Irene Cantarero‐Villanueva, George David Baxter, Acute and cumulative benefits of Photobiomodulation for xerostomia: A systematic review and meta‐analysis, 2020, 1354-523X, 10.1111/odi.13648 | |
173. | Leila Gholami, Sohrab Asefi, Amirarsalan Hooshyarfard, Anton Sculean, Georgios E. Romanos, Akira Aoki, Reza Fekrazad, Photobiomodulation in Periodontology and Implant Dentistry: Part 1, 2019, 37, 2578-5478, 739, 10.1089/photob.2019.4710 | |
174. | Evaldo Moreira da Silva, Raquel Agnelli Mesquita-Ferrari, Maria Fernanda Setúbal Destro Rodrigues, Eric Moreno Ramos Magalhães, Sandra Kalil Bussadori, Adriana De Brito, Felipe de Moura Messias, Nadhia Helena Costa Souza, Agnelo Neves Alves, Kristianne Porta Santos Fernandes, The Effects of Photobiomodulation on Inflammatory Infiltrate During Muscle Repair in Advanced-Age Rats, 2019, 1079-5006, 10.1093/gerona/glz076 | |
175. | Ramón Iglesias-Rey, José Castillo, New strategies for ischemic stroke: internal photobiomodulation therapy, 2020, 15, 1673-5374, 1658, 10.4103/1673-5374.276328 | |
176. | Andrezza Maria Côrtes Thomé Lima, Luiz Philippe da Silva Sergio, Adenilson de Souza da Fonseca, Photobiomodulation via multiple-wavelength radiations, 2020, 35, 0268-8921, 307, 10.1007/s10103-019-02879-1 | |
177. | Association of Bioglass/Collagen/Magnesium composites and low level irradiation: effects on bone healing in a model of tibial defect in rats, 2018, 27, 0898-5901, 271, 10.5978/islsm.27_18-OR-25 | |
178. | Patricia Gabielli Vassão, Julia Parisi, Thaíse Fernanda Campos Penha, Ana Beatriz Balão, Ana Claudia Muniz Renno, Mariana Arias Avila, Association of photobiomodulation therapy (PBMT) and exercises programs in pain and functional capacity of patients with knee osteoarthritis (KOA): a systematic review of randomized trials, 2021, 0268-8921, 10.1007/s10103-020-03223-8 | |
179. | Kaline de Brito Sousa, Daniela de Fátima Teixeira da Silva, Maria Fernanda Setúbal Destro Rodrigues, Mónica Pereira Garcia, Carolina de Oliveira Rodini, Raquel Agnelli Mesquita‐Ferrari, Michael R Hamblin, Sandra Kalil Bussadori, Fabio Daumas Nunes, Kristianne Porta Santos Fernandes, Effects of the phenotypic polarization state of human leukocytes on the optical absorbance spectrum, 2021, 1864-063X, 10.1002/jbio.202000487 | |
180. | Gurbind Singh, Divya Sridharan, Mahmood Khan, Polani B. Seshagiri, Mouse embryonic stem cell-derived cardiomyocytes cease to beat following exposure to monochromatic light: association with increased ROS and loss of calcium transients, 2019, 317, 0363-6143, C725, 10.1152/ajpcell.00188.2019 | |
181. | Katia Llanos do Vale, Durvanei Augusto Maria, Lara Cristina Picoli, Alessandro Melo Deana, Marcelo Betti Mascaro, Raquel Agnelli Mesquita Ferrari, Sandra Kalil Bussadori, Kristianne Porta Santos Fernandes, The Effects of Photobiomodulation Delivered by Light-Emitting Diode on Stem Cells from Human Exfoliated Deciduous Teeth: A Study on the Relevance to Pluripotent Stem Cell Viability and Proliferation, 2017, 35, 1557-8550, 659, 10.1089/pho.2017.4279 | |
182. | Camila Ayumi Ivanaga, Daniela Maria Janjacomo Miessi, Marta Aparecida Alberton Nuernberg, Marina Módolo Claudio, Valdir Gouveia Garcia, Leticia Helena Theodoro, Antimicrobial photodynamic therapy (aPDT) with curcumin and LED, as an enhancement to scaling and root planing in the treatment of residual pockets in diabetic patients: A randomized and controlled split-mouth clinical trial, 2019, 27, 15721000, 388, 10.1016/j.pdpdt.2019.07.005 | |
183. | Shing Yau Tam, Victor C. W. Tam, Shanmugasundaram Ramkumar, May Ling Khaw, Helen K. W. Law, Shara W. Y. Lee, Review on the Cellular Mechanisms of Low-Level Laser Therapy Use in Oncology, 2020, 10, 2234-943X, 10.3389/fonc.2020.01255 | |
184. | Marcelo Frigero, Solange Almeida dos Santos, Andrey Jorge Serra, Caroline dos Santos Monteiro Machado, Leslie Andrews Portes, Paulo José Ferreira Tucci, Flavio Silva, Ernesto Cesar Leal-Junior, Paulo de Tarso Camillo de Carvalho, Effect of photobiomodulation therapy on oxidative stress markers of gastrocnemius muscle of diabetic rats subjected to high-intensity exercise, 2018, 33, 0268-8921, 1781, 10.1007/s10103-018-2540-7 | |
185. | Ruwaidah A. Mussttaf, David F. L. Jenkins, Awadhesh N. Jha, Assessing the impact of low level laser therapy (LLLT) on biological systems: a review, 2019, 95, 0955-3002, 120, 10.1080/09553002.2019.1524944 | |
186. | Maria Francesca Sfondrini, Marina Vitale, Antonio Luiz Barbosa Pinheiro, Paola Gandini, Lorenzo Sorrentino, Ugo Matteo Iarussi, Andrea Scribante, Photobiomodulation and Pain Reduction in Patients Requiring Orthodontic Band Application: Randomized Clinical Trial, 2020, 2020, 2314-6133, 1, 10.1155/2020/7460938 | |
187. | Armin Khosravipour, Abdollah Amini, Reza Masteri Farahani, Fatemeh Zare, Atarodsadat Mostafavinia, Somaye Fallahnezhad, Saman Akbarzade, Mehrdad Asgari, Ahmad Mohammadbeigi, Fatemehsadat Rezaei, Seyed Kamran Ghoreishi, Sufan Chien, Mohammad Bayat, Preconditioning adipose-derived stem cells with photobiomodulation significantly increased bone healing in a critical size femoral defect in rats, 2020, 531, 0006291X, 105, 10.1016/j.bbrc.2020.07.048 | |
188. | Prasun Chatterjee, Achal K. Srivastava, Deepa A. Kumar, Avinash Chakrawarty, Maroof A. Khan, Akash K. Ambashtha, Vijay Kumar, Luis De Taboada, Aparajit B. Dey, Effect of deep tissue laser therapy treatment on peripheral neuropathic pain in older adults with type 2 diabetes: a pilot randomized clinical trial, 2019, 19, 1471-2318, 10.1186/s12877-019-1237-5 | |
189. | Francesca Maria Salmeri, Lucia Denaro, Elisa Ruello, Giuseppe Acri, Sergio Gurgone, Carlo Sansotta, Barbara Testagrossa, Irradiation with Polychromatic Incoherent Low Energy Radiation of Human Peripheral Blood Mononuclear Cells In Vitro: Effects on Cytokine Production, 2020, 17, 1660-4601, 1233, 10.3390/ijerph17041233 | |
190. | Reem Hanna, Snehal Dalvi, Tudor Sălăgean, Ioana Delia Pop, Ioana Roxana Bordea, Stefano Benedicenti, Understanding COVID-19 Pandemic: Molecular Mechanisms and Potential Therapeutic Strategies. An Evidence-Based Review, 2021, Volume 14, 1178-7031, 13, 10.2147/JIR.S282213 | |
191. | Xuemei Zong, Yan Dong, Yuyu Li, Luodan Yang, Yong Li, Baocheng Yang, Lorelei Tucker, Ningjun Zhao, Darrell W. Brann, Xianliang Yan, Shuqun Hu, Quanguang Zhang, Beneficial Effects of Theta-Burst Transcranial Magnetic Stimulation on Stroke Injury via Improving Neuronal Microenvironment and Mitochondrial Integrity, 2020, 11, 1868-4483, 450, 10.1007/s12975-019-00731-w | |
192. | Ralph Fernando Rosas, Aline Armiliato Emer, Ana Paula Batisti, Daniela Dero Ludtke, Bruna Lenfers Turnes, Franciane Bobinski, Francisco José Cidral-Filho, Daniel Fernandes Martins, Far infrared-emitting ceramics decrease Freund’s adjuvant-induced inflammatory hyperalgesia in mice through cytokine modulation and activation of peripheral inhibitory neuroreceptors, 2018, 16, 20954964, 396, 10.1016/j.joim.2018.08.002 | |
193. | Andrezza Maria Côrtes Thomé Lima, Luiz Philippe da Silva Sergio, Larissa Alexsandra da Silva Neto Trajano, Bianca Paschoal de Souza, João Pedro da Motta Mendes, Adriano Fonseca Ribeiro Cardoso, Caroline Pisa Figueira, Beatriz dos Anjos Tavares, Daniella Sousa Figueira, André Luiz Mencalha, Eduardo Tavares Lima Trajano, Adenilson de Souza da Fonseca, Photobiomodulation by dual-wavelength low-power laser effects on infected pressure ulcers, 2020, 35, 0268-8921, 651, 10.1007/s10103-019-02862-w | |
194. | Patricia Gabrielli Vassão, Ana Claudia Renno, Bjorn N. Smith, Gregory B. Bennett, Michael Murphy, Ann Liebert, Roberta Chow, E-Liisa Laakso, Pre-Conditioning and Post-Operative Photobiomodulation Therapy by a Novel Light Patch System for Knee Arthroplasty: A Protocol for a Phase 1 Study, 2020, 38, 2578-5478, 206, 10.1089/photob.2019.4751 | |
195. | Giovanna Moura Lamas Della Santa, Marcílio Coelho Ferreira, Thaís Peixoto Gaiad Machado, Murilo Xavier Oliveira, Ana Paula Santos, Effects of Photobiomodulation Therapy (LED 630 nm) on Muscle and Nerve Histomorphometry after Axonotmesis, 2021, 0031-8655, 10.1111/php.13415 | |
196. | Melissa J. Lewis, Nicolas Granger, Nick D. Jeffery, Emerging and Adjunctive Therapies for Spinal Cord Injury Following Acute Canine Intervertebral Disc Herniation, 2020, 7, 2297-1769, 10.3389/fvets.2020.579933 | |
197. | Burkhard T. Espey, Karin Kielwein, Hans Ven, Klaus Steger, Jean‐Pierre Allam, Agnieszka Paradowska‐Dogan, Katrin Ven, Effects of Pulsed‐Wave Photobiomodulation Therapy on Human Spermatozoa, 2021, 0196-8092, 10.1002/lsm.23399 | |
198. | Snehal Dalvi, Namrata Khetal, Salman Ansari, Stefano Benedicenti, Reem Hanna, Utilization of 810 nm Diode Laser Treatment in Periodontitis as an Alternative to Surgical Debridement Approach, 2021, 0031-8655, 10.1111/php.13417 | |
199. | Eduardo Quintão Manhanini Souza, Tiago Esgalha da Rocha, Luan Felipe Toro, Isabella Zacarin Guiati, Jéssica de Oliveira Alvarenga Freire, Edilson Ervolino, Daniela Atili Brandini de Weert, Valdir Gouveia Garcia, Letícia Helena Theodoro, Adjuvant Effects of Curcumin as a Photoantimicrobial or Irrigant in the Non-surgical Treatment of Periodontitis: Systematic Review and Meta-Analysis, 2021, 15721000, 102265, 10.1016/j.pdpdt.2021.102265 | |
200. | Fatereh Rezaei, Mohammad Bayat, Hamid Nazarian, Abbas Aliaghaei, Hojjat-Allah Abaszadeh, Parvaneh Naserzadeh, Abdollah Amini, Vahid Ebrahimi, Shabnam Abdi, Mohammad-Amin Abdollahifar, Photobiomodulation Therapy Improves Spermatogenesis in Busulfan-Induced Infertile Mouse, 2021, 1933-7191, 10.1007/s43032-021-00557-8 | |
201. | Xiaotong Ren, Yilin Chou, Yuexin Wang, Yanyan Chen, Ziyuan Liu, Xuemin Li, Comparison of intense pulsed light and near‐infrared light in the treatment of dry eye disease: a prospective randomized study, 2021, 1755-375X, 10.1111/aos.14833 | |
202. | Farzad Salehpour, Mahsa Khademi, Michael R. Hamblin, Photobiomodulation Therapy for Dementia: A Systematic Review of Pre-Clinical and Clinical Studies, 2021, 13872877, 1, 10.3233/JAD-210029 | |
203. | Willians Fernando Vieira, Bruno Kenzo-Kagawa, Lúcia Elvira Alvares, José Carlos Cogo, Vitor Baranauskas, Maria Alice da Cruz-Höfling, Exploring the ability of low-level laser irradiation to reduce myonecrosis and increase Myogenin transcription after Bothrops jararacussu envenomation, 2021, 20, 1474-905X, 571, 10.1007/s43630-021-00041-x | |
204. | Danielle da Silva Pompeu, Brennda Lucy Freitas de Paula, Antônia Patricia Oliveira Barros, Samir Costa Nunes, Alexandra Melo Pingarilho Carneiro, Jesuína Lamartine Nogueira Araújo, Cecy Martins Silva, Alessandro Loguercio, Combination of strontium chloride and photobiomodulation in the control of tooth sensitivity post-bleaching: A split-mouth randomized clinical trial, 2021, 16, 1932-6203, e0250501, 10.1371/journal.pone.0250501 | |
205. | Marootpong Pooam, Blanche Aguida, Soria Drahy, Nathalie Jourdan, Margaret Ahmad, Therapeutic application of light and electromagnetic fields to reduce hyper-inflammation triggered by COVID-19, 2021, 14, 1942-0889, 66, 10.1080/19420889.2021.1911413 | |
206. | Renlong Zhang, Ting Zhou, Liwei Liu, Tymish Y Ohulchanskyy, Junle Qu, Dose–effect relationships for PBM in the treatment of Alzheimer’s disease, 2021, 54, 0022-3727, 353001, 10.1088/1361-6463/ac0740 | |
207. | Tereza Cristina Buzinari, Thiago Francisco de Moraes, Julio Cesar Conceição-Filho, Evelin Capellari Cárnio, Luciana Almeida-Lopes, Helio Cesar Salgado, Gerson Jhonatan Rodrigues, Nitric oxide storage levels modulate vasodilation and the hypotensive effect induced by photobiomodulation using an aluminum gallium arsenide (AlGaAs) diode laser (660 nm), 2022, 37, 1435-604X, 2753, 10.1007/s10103-022-03551-x | |
208. | Han-Joon Kim, Weni Sritandi, Ze Xiong, John S. Ho, Bioelectronic devices for light-based diagnostics and therapies, 2023, 4, 2688-4089, 011304, 10.1063/5.0102811 | |
209. | María Roxana Ferreira Sertaje, Marcela Leticia Leal Gonçalves, Andréa Oliver Gomes, Laura Hermida Bruno, Ana Laura Fossati, Natalia Osorio Viarengo, Elaine Marcilio Santos, Ana Paula Taboada Sobral, Raquel Agnelli Mesquita-Ferrari, Kristianne Porta Santos Fernandes, Anna Carolina Ratto Tempestini Horliana, Lara Jansiski Motta, Sandra Kalil Bussadori, Vascular photobiomodulation in the treatment of children with temporomandibular disorders: Study protocol for a randomized, controlled, blind, clinical trial, 2022, 101, 1536-5964, e31228, 10.1097/MD.0000000000031228 | |
210. | Reza Fekrazad, Sohrab Asefi, Maryam Pourhajibagher, Farshid Vahdatinia, Sepehr Fekrazad, Abbas Bahador, Heidi Abrahamse, Michael R. Hamblin, 2021, Chapter 30, 978-3-030-63760-6, 517, 10.1007/978-3-030-63761-3_30 | |
211. | Claudiane Pedro Rodrigues, Jeferson Lucas Jacinto, Mirela Casonato Roveratti, Jeanne Karlette Merlo, Lúcio Flávio Soares-Caldeira, Alex Silva Ribeiro, João Pedro Nunes, Eros de Oliveira Junior, Andreo Fernando Aguiar, Effects of Photobiomodulation/Laser Therapy Combined With Resistance Training on Quadriceps Hypertrophy and Strength, and Postural Balance in Older Women: A Randomized, Triple-Blinded, Placebo-Controlled Study, 2022, 45, 1539-8412, 125, 10.1519/JPT.0000000000000313 | |
212. | Pedro Paulo Ribeiro Ferreira, Luís Filipe Rocha Silva, Marco Fabrício Dias-Peixoto, Ricardo Cardoso Cassilhas, Fernando Gripp, Fabiano Trigueiro Amorim, Zachary A. Mang, Elizabethe Adriana Esteves, Valmor A. Tricoli, Cleber Ferraresi, Flávio de Castro Magalhães, Effects of the association of different volumes of strength training with photobiomodulation therapy on insulin resistance: A protocol for a randomized, triple-blind, placebo-controlled trial, 2022, 29, 24518654, 100984, 10.1016/j.conctc.2022.100984 | |
213. | Iuliia Golovynska, Yurii V. Stepanov, Sergii Golovynskyi, Ting Zhou, Liudmyla I. Stepanova, Liudmyla V. Garmanchuk, Tymish Y. Ohulchanskyy, Junle Qu, Macrophages Modulated by Red/NIR Light: Phagocytosis, Cytokines, Mitochondrial Activity, Ca 2+ Influx, Membrane Depolarization and Viability , 2022, 98, 0031-8655, 484, 10.1111/php.13526 | |
214. | Steven Parker, Mark Cronshaw, Martin Grootveld, Photobiomodulation Delivery Parameters in Dentistry: An Evidence-Based Approach, 2022, 40, 2578-5478, 42, 10.1089/photob.2021.0116 | |
215. | Zhijie Zhu, Xuankang Wang, Zhiwen Song, Xiaoshuang Zuo, Yangguang Ma, Zhihao Zhang, Cheng Ju, Zhuowen Liang, Kun Li, Xueyu Hu, Zhe Wang, Photobiomodulation promotes repair following spinal cord injury by restoring neuronal mitochondrial bioenergetics via AMPK/PGC-1α/TFAM pathway, 2022, 13, 1663-9812, 10.3389/fphar.2022.991421 | |
216. | Michael R. Hamblin, 2022, Chapter 23, 978-3-030-92879-7, 273, 10.1007/978-3-030-92880-3_23 | |
217. | Marina Curra, Julianna Joanna Carvalho Moraes de Campos Baldin, Marco Antonio Trevizani Martins, Lauren Frenzel Schuch, Ana Luísa Saraiva Homem de Carvalho, Eduardo José Gaio, Cassiano Kuchenbecker Rösing, Rosane Isabel Bittencourt, Lauro José Gregianin, Alessandra Aparecida Paz, Liane Esteves Daudt, Manoela Domingues Martins, Investigation of oral and general health status and IL-1β gene polymorphism as risk factors for oral mucositis in hematopoietic stem cell transplantation patients, 2022, 36, 1807-3107, 10.1590/1807-3107bor-2022.vol36.0016 | |
218. | Reiza Dolendo Ventura, A review on photobiomodulation therapy for olfactory dysfunction caused by COVID-19, 2022, 11, 2287-8300, 72, 10.25289/ML.2022.11.2.72 | |
219. | Laura Ailioaie, Gerhard Litscher, Probiotics, Photobiomodulation, and Disease Management: Controversies and Challenges, 2021, 22, 1422-0067, 4942, 10.3390/ijms22094942 | |
220. | Giada Magni, Domenico Piccolo, Paolo Bonan, Claudio Conforti, Giuliana Crisman, Laura Pieri, Irene Fusco, Francesca Rossi, 1540-nm fractional laser treatment modulates proliferation and neocollagenesis in cultured human dermal fibroblasts, 2022, 9, 2296-858X, 10.3389/fmed.2022.1010878 | |
221. | Tomoyasu Yamanaka, Takatoshi Ueki, Mitsuhito Mase, Koichi Inoue, Arbitrary Ca2+ regulation for endothelial nitric oxide, NFAT and NF-κB activities by an optogenetic approach, 2023, 13, 1663-9812, 10.3389/fphar.2022.1076116 | |
222. | Marcos Aurélio Teixeira, Juliana Zampoli Boava Papini, Aguinaldo Garcez, Giovana Radomille Tofoli, Comparative analysis of two laser wavelengths in the stimulation of acupuncture points for analgesic effects in an animal model, 2022, 15, 1864-063X, 10.1002/jbio.202100213 | |
223. | Sara Pourshahidi, Hooman Ebrahimi, Naghmeh Bahrami, Zahra Abbasi Javan, Younes Ghoreyshi, Nasim Chiniforush, Mohammad Javad Kharazifard, In vitro Effect of 810 nm and 940 nm Diode Laser Irradiation on Proliferation of Human Gingival Fibroblasts and Expression of Procollagen Gene , 2022, 98, 0031-8655, 1441, 10.1111/php.13630 | |
224. | Débora Buendía, Tatiana Guncay, Macarena Oyanedel, Makarena Lemus, Alejandro Weinstein, Álvaro O. Ardiles, José Marcos, Adriana Fernandes, Renato Zângaro, Pablo Muñoz, The Transcranial Light Therapy Improves Synaptic Plasticity in the Alzheimer’s Disease Mouse Model, 2022, 12, 2076-3425, 1272, 10.3390/brainsci12101272 | |
225. | Seyyedeh Niloufar Tabatabaei, Mahshid Hodjat, Neda Hakimiha, Mohammad Sadegh Ahmad Akhoundi, Mohammad Javad Kharazifard, In Vitro Effect of Photobiomodulation Therapy with 980 nm Diode Laser on Gene Expression of Key Regulators of Bone Remodeling by Human Periodontal Ligament Cells under Mild Orthodontic Forces , 2023, 0031-8655, 10.1111/php.13787 | |
226. | Andrea Marchegiani, Alessandro Fruganti, Marilena Bazzano, Matteo Cerquetella, Fabrizio Dini, Andrea Spaterna, Fluorescent Light Energy in the Management of Multi Drug Resistant Canine Pyoderma: A Prospective Exploratory Study, 2022, 11, 2076-0817, 1197, 10.3390/pathogens11101197 | |
227. | Ha Thanh-Thai Le, Nam Cong-Nhat Huynh, Quynh-Anh Nguyen-Ho, Thuy Thu Nguyen, Son Hoang Le, Ly Thi-Bich Nguyen, Effect of Photobiomodulation Therapy on Reducing Acute Pain and Inflammation Following Surgical Removal of Impacted Mandibular Third Molars: A Randomized, Split-Mouth Clinical Trial, 2022, 40, 2578-5478, 245, 10.1089/photob.2021.0110 | |
228. | Renan Vieira de Brito, Marília Wellichan Mancini, Marcel das Neves Palumbo, Luis Henrique Oliveira de Moraes, Gerson Jhonatan Rodrigues, Onivaldo Cervantes, Joel Avram Sercarz, Marcos Bandiera Paiva, The Rationale for “Laser-Induced Thermal Therapy (LITT) and Intratumoral Cisplatin” Approach for Cancer Treatment, 2022, 23, 1422-0067, 5934, 10.3390/ijms23115934 | |
229. | A. O. Maslakova, M. Ya. Liuta, N. O. Sybirna, Effect of photobiomodulation therapy on the regulation of glucose uptake by lymphocytes in diabetes mellitus (Review), 2021, 15, 19964536, 87, 10.30970/sbi.1504.671 | |
230. | Kangzhe Xie, Hala El Khoury, John Mitrofanis, Paul J. Austin, A systematic review of the effect of photobiomodulation on the neuroinflammatory response in animal models of neurodegenerative diseases, 2022, 0, 0334-1763, 10.1515/revneuro-2022-0109 | |
231. | In-Young Jo, Hyung-Kwon Byeon, Myung-Jin Ban, Jae-Hong Park, Sang-Cheol Lee, Young-Kyun Won, Yun-Su Eun, Jae-Yun Kim, Na-Gyeong Yang, Sul-Hee Lee, Pyeongan Lee, Nam-Hun Heo, Sujin Jo, Hoonhee Seo, Sukyung Kim, Ho-Yeon Song, Jung-Eun Kim, Effect of a Novel Handheld Photobiomodulation Therapy Device in the Management of Chemoradiation Therapy-Induced Oral Mucositis in Head and Neck Cancer Patients: A Case Series Study, 2023, 10, 2304-6732, 241, 10.3390/photonics10030241 | |
232. | Xiaohui Zhao, Wanyi Tang, Haipeng Wang, Hao He, Femtosecond-laser stimulation induces senescence of tumor cells in vitro and in vivo, 2022, 13, 2156-7085, 791, 10.1364/BOE.449456 | |
233. | K S Canuto, I S S Amorim, J A Rodrigues, A F Teixeira, A L Mencalha, A S Fonseca, Effects of photobiomodulation by low power lasers on the in vitro proliferation and aggressiveness of breast cancer cells, 2021, 31, 1054-660X, 085603, 10.1088/1555-6611/ac0980 | |
234. | ChandrashekarMurugesh Yavagal, Akshaya Lal, ViplaviVijaysinh Chavan Patil, PujaC Yavagal, KiranKumar Neelakantappa, Madhu Hariharan, Efficacy of laser photobiomodulation pulpotomy in human primary teeth: A randomized controlled trial, 2021, 39, 0970-4388, 436, 10.4103/jisppd.jisppd_150_21 | |
235. | Tomasz Walski, Karolina Grzeszczuk-Kuć, Katarzyna Gałecka, Natalia Trochanowska-Pauk, Raghvendra Bohara, Albert Czerski, Konstanty Szułdrzyński, Wiesław Królikowski, Jerzy Detyna, Małgorzata Komorowska, Near-infrared photobiomodulation of blood reversibly inhibits platelet reactivity and reduces hemolysis, 2022, 12, 2045-2322, 10.1038/s41598-022-08053-y | |
236. | Shiyao Xu, Bo Wan, Recent advances in low-level laser therapy on depression, 2022, 2, 2709-1325, 123, 10.26599/SAB.2022.9060026 | |
237. | Yul Hee Kim, Hyeung Kyoo Kim, Jee Woong Choi, You Chan Kim, Photobiomodulation therapy with an 830-nm light-emitting diode for the prevention of thyroidectomy scars: a randomized, double-blind, sham device-controlled clinical trial, 2022, 37, 1435-604X, 3583, 10.1007/s10103-022-03637-6 | |
238. | Henrique Hadad, Ana Flávia Piquera Santos, Laís Kawamata de Jesus, Pier Paolo Poli, Ronaldo Célio Mariano, Leticia Helena Theodoro, Carlo Maiorana, Francisley Ávila Souza, Photobiomodulation Therapy Improves Postoperative Pain and Edema in Third Molar Surgeries: A Randomized, Comparative, Double-Blind, and Prospective Clinical Trial, 2022, 80, 02782391, 37.e1, 10.1016/j.joms.2021.08.267 | |
239. | Atul Dhall, Jun Ying Tan, Min Jun Oh, Sayemul Islam, Jungkwun Kim, Albert Kim, Geelsu Hwang, A dental implant-on-a-chip for 3D modeling of host–material–pathogen interactions and therapeutic testing platforms, 2022, 22, 1473-0197, 4905, 10.1039/D2LC00774F | |
240. | Woori Choi, Ku Youn Baik, Seung Jeong, Sangbae Park, Jae Eun Kim, Hong Bae Kim, Jong Hoon Chung, Photobiomodulation as an antioxidant substitute in post-thawing trauma of human stem cells from the apical papilla, 2021, 11, 2045-2322, 10.1038/s41598-021-96841-3 | |
241. | A.C. Barolet, I.V. Litvinov, D. Barolet, Light-induced nitric oxide release in the skin beyond UVA and blue light: Red & near-infrared wavelengths, 2021, 117, 10898603, 16, 10.1016/j.niox.2021.09.003 | |
242. | Sang Ho Lee, Yu-Jin Kim, Yeong Hwan Kim, Han Young Kim, Suk Ho Bhang, Enhancing therapeutic efficacy of human adipose-derived stem cells by modulating photoreceptor expression for advanced wound healing, 2022, 13, 1757-6512, 10.1186/s13287-022-02892-2 | |
243. | Gokhan Sert, Arda Kucukguven, Elham Bahador Zırh, Tugrul T. Demirtaş, Ayşe Nur Çakar, Menemşe Gümüşderelioğlu, Mert Calis, Photobiomodulation with polychromatic light (600–1200 nm) improves fat graft survival by increasing adipocyte viability, neovascularization, and reducing inflammation in a rat model, 2022, 54, 0196-8092, 268, 10.1002/lsm.23457 | |
244. | Robson Alexandre Brochetti, Simone Klein, Paula Tatiane Alonso, Adriana Schapochnik, Amílcar Sabino Damazo, Michael R. Hamblin, Maria Fernanda de Souza Setubal Destro, Adriana Lino-dos-Santos-Franco, Beneficial effects of infrared light-emitting diode in corticosteroid-resistant asthma, 2022, 37, 1435-604X, 1963, 10.1007/s10103-021-03457-0 | |
245. | M. Sadraeian, Le Zhang, F. Aavani, E. Biazar, D. Jin, Photodynamic viral inactivation assisted by photosensitizers, 2022, 28, 25425293, 100882, 10.1016/j.mtphys.2022.100882 | |
246. | Rolando Toyos, Neel R. Desai, Melissa Toyos, Steven J. Dell, Walid Kamal Abdelbasset, Intense pulsed light improves signs and symptoms of dry eye disease due to meibomian gland dysfunction: A randomized controlled study, 2022, 17, 1932-6203, e0270268, 10.1371/journal.pone.0270268 | |
247. | Ji Soo Baik, Tae Young Lee, Nam Gyun Kim, Kyoungjune Pak, Sung-Hwa Ko, Ji Hong Min, Yong-Il Shin, Effects of Photobiomodulation on Changes in Cognitive Function and Regional Cerebral Blood Flow in Patients with Mild Cognitive Impairment: A Pilot Uncontrolled Trial, 2021, 83, 13872877, 1513, 10.3233/JAD-210386 | |
248. | Letícia Helena THEODORO, Rosemary Adriana Chiérici MARCANTONIO, Mark WAINWRIGHT, Valdir Gouveia GARCIA, LASER in periodontal treatment: is it an effective treatment or science fiction?, 2021, 35, 1807-3107, 10.1590/1807-3107bor-2021.vol35.0099 | |
249. | Heidi Abrahamse, Photobiomodulation and Oxidative/Nitrosative Stress, 2021, 39, 2578-5478, 685, 10.1089/photob.2021.0084 | |
250. | Brenda Thaynne Lima de Matos, Daniela Vieira Buchaim, Karina Torres Pomini, Sandra Maria Barbalho, Elen Landgraf Guiguer, Carlos Henrique Bertoni Reis, Cleuber Rodrigo de Souza Bueno, Marcelo Rodrigues da Cunha, Eliana de Souza Bastos Mazuqueli Pereira, Rogerio Leone Buchaim, Photobiomodulation Therapy as a Possible New Approach in COVID-19: A Systematic Review, 2021, 11, 2075-1729, 580, 10.3390/life11060580 | |
251. | Camila Amaral Coracini, Francyelle dos Santos Soares, Gladson Ricardo Flor Bertolini, The use of LED therapy to treat synovial joints disorders: scoping review, 2022, 37, 0268-8921, 701, 10.1007/s10103-021-03372-4 | |
252. | Zhuowen Liang, Tao Lei, Shuang Wang, Pan Li, Beiyu Chen, Dongsheng Pan, Yongfeng Zhang, Xiaoshuang Zuo, Xuankang Wang, Zhuojing Luo, Xueyu Hu, Tan Ding, Zhe Wang, Clinical safety study of photobiomodulation in acute spinal cord injury by scattering fiber, 2022, 37, 1435-604X, 3433, 10.1007/s10103-022-03601-4 | |
253. | Xiaobin Wu, Jongho Park, Siu Yu A. Chow, Maria Carmelita Z. Kasuya, Yoshiho Ikeuchi, Beomjoon Kim, Localised light delivery on melanoma cells using optical microneedles, 2022, 13, 2156-7085, 1045, 10.1364/BOE.450456 | |
254. | Yoo-Seung Ko, Eun-Ji Gi, Sungsu Lee, Hyong-Ho Cho, Dual red and near-infrared light-emitting diode irradiation ameliorates LPS-induced otitis media in a rat model, 2023, 11, 2296-4185, 10.3389/fbioe.2023.1099574 | |
255. | Nicholas Tripodi, Fotios Sidiroglou, Sarah Fraser, Maja Husaric, Dimitrios Kiatos, Vasso Apostolopoulos, Jack Feehan, The effects of polarized photobiomodulation on cellular viability, proliferation, mitochondrial membrane potential and apoptosis in human fibroblasts: Potential applications to wound healing, 2022, 236, 10111344, 112574, 10.1016/j.jphotobiol.2022.112574 | |
256. | S. Salman, C. Guermonprez, L. Declercq, S. Kerdine-Römer, P05-03 Photobiomodulation-induced Nrf2 partially controls the inflammatory response in keratinocytes, 2022, 368, 03784274, S109, 10.1016/j.toxlet.2022.07.312 | |
257. | Sihan Guo, Ruimin Wang, Jiewei Hu, Liping Sun, Xinru Zhao, Yufeng Zhao, Dong Han, Shuqun Hu, Photobiomodulation Promotes Hippocampal CA1 NSC Differentiation Toward Neurons and Facilitates Cognitive Function Recovery Involving NLRP3 Inflammasome Mitigation Following Global Cerebral Ischemia, 2021, 15, 1662-5102, 10.3389/fncel.2021.731855 | |
258. | Jie Ding, Maiken Mellergaard, Zhensen Zhu, Peter Kwan, Deirdre Edge, Zengshuan Ma, Lise Hebert, Saad Alrobaiea, Takashi Iwasaki, Michael Canova Engelbrecht Nielsen, Edward E. Tredget, Fluorescent light energy modulates healing in skin grafted mouse model, 2021, 16, 2391-5463, 1240, 10.1515/med-2021-0329 | |
259. | Imran Khan, Saeed Ur Rahman, Elieza Tang, Karl Engel, Bradford Hall, Ashok B. Kulkarni, Praveen R. Arany, Accelerated burn wound healing with photobiomodulation therapy involves activation of endogenous latent TGF-β1, 2021, 11, 2045-2322, 10.1038/s41598-021-92650-w | |
260. | Jolien Robijns, Raj G. Nair, Joy Lodewijckx, Praveen Arany, Andrei Barasch, Jan M. Bjordal, Paolo Bossi, Anne Chilles, Patricia M. Corby, Joel B. Epstein, Sharon Elad, Reza Fekrazad, Eduardo Rodrigues Fregnani, Marie-Thérèse Genot, Ana M. C. Ibarra, Michael R. Hamblin, Vladimir Heiskanen, Ken Hu, Jean Klastersky, Rajesh Lalla, Sofia Latifian, Arun Maiya, Jeroen Mebis, Cesar A. Migliorati, Dan M. J. Milstein, Barbara Murphy, Judith E. Raber-Durlacher, Hendrik J. Roseboom, Stephen Sonis, Nathaniel Treister, Yehuda Zadik, René-Jean Bensadoun, Photobiomodulation therapy in management of cancer therapy-induced side effects: WALT position paper 2022, 2022, 12, 2234-943X, 10.3389/fonc.2022.927685 | |
261. | Flávio de Castro Magalhães, Cleber Ferraresi, Photobiomodulation Therapy on the Treatment of Insulin Resistance: A Narrative Review, 2022, 40, 2578-5478, 597, 10.1089/photob.2022.0031 | |
262. | Ricardo Henrique Esquivel Azuma, Jeanne Karlette Merlo, Jeferson Lucas Jacinto, Jayne Maria Borim, Rubens Alexandre da Silva, Francis Lopes Pacagnelli, Joao Pedro Nunes, Alex Silva Ribeiro, Andreo Fernando Aguiar, Photobiomodulation Therapy at 808 nm Does Not Improve Biceps Brachii Performance to Exhaustion and Delayed-Onset Muscle Soreness in Young Adult Women: A Randomized, Controlled, Crossover Trial, 2021, 12, 1664-042X, 10.3389/fphys.2021.664582 | |
263. | Mirian Bonifacio, Izabelle Dias Benfato, Matheus de Almeida Cruz, Daniele Correia de Sales, Isabella Liba Pandolfo, Hananiah Tardivo Quintana, Carolina Prado de França Carvalho, Camila Aparecida Machado de Oliveira, Ana Cláudia Muniz Renno, Effects of photobiomodulation on glucose homeostasis and morphometric parameters in pancreatic islets of diabetic mice, 2022, 37, 1435-604X, 1799, 10.1007/s10103-021-03434-7 | |
264. | Jéssica Iohanna da Silva, Ana Larysa Galdino das Chagas, Bárbara de Oliveira Sena, Camila Almeida de Lima, Giulia Viana dos Santos, Maria Carolina Dantas Campelo, Lays Pinheiro de Medeiros, Rhayssa de Oliveira e Araújo, Intervenções eficazes para tratamento de trauma mamilar decorrente da amamentação: revisão sistemática, 2022, 35, 0103-2100, 10.37689/acta-ape/2022AR0001367 | |
265. | Zeqing Chen, Ruixiao Zhang, Haokuan Qin, Hui Jiang, Aixia Wang, Xiaolin Zhang, Shijie Huang, Miao Sun, Xuewei Fan, Zhicheng Lu, Yinghua Li, Shangfeng Liu, Muqing Liu, The pulse light mode enhances the effect of photobiomodulation on B16F10 melanoma cells through autophagy pathway, 2023, 38, 1435-604X, 10.1007/s10103-023-03733-1 | |
266. | Yi-Chuan Chang, Chun-Ming Chen, Ing-Shiow Lay, Yu-Chen Lee, Cheng-Hao Tu, The Dosage Effect of Laser Acupuncture at PC6 (Neiguan) on Heart Rate Variability: A Pilot Study, 2022, 12, 2075-1729, 1951, 10.3390/life12121951 | |
267. | Ralph Zipper, Georgine Lamvu, Vaginal laser therapy for gynecologic conditions: re-examining the controversy and where do we go from here, 2022, 11, 2042-6305, 843, 10.2217/cer-2021-0281 | |
268. | Brian Bicknell, Ann Liebert, Craig S. McLachlan, Hosen Kiat, Microbiome Changes in Humans with Parkinson’s Disease after Photobiomodulation Therapy: A Retrospective Study, 2022, 12, 2075-4426, 49, 10.3390/jpm12010049 | |
269. | Hyeong Ju Park, Yeon Hee Cho, Hyeyoon Goo, Sun-hyang Choi, Eun Young Kim, Namgue Hong, SangJoon Mo, Ji On Park, Byeong-Il Lee, Min Young Lee, Jin-Chul Ahn, Emiliano Antiga, Treatment with Light-Emitting Diodes of Wavelength 863 nm Delays DMBA/TPA-Induced Skin Tumor Formation and Decreases Proinflammatory Cytokine Levels in ICR Mice, 2022, 2022, 2314-6141, 1, 10.1155/2022/4400276 | |
270. | Atarodsadat Mostafavinia, Houssein Ahmadi, Abdollah Amini, Zahra Roudafshani, Michael R Hamblin, Sufan Chien, Mohammad Bayat, The effect of photobiomodulation therapy on antioxidants and oxidative stress profiles of adipose derived mesenchymal stem cells in diabetic rats, 2021, 262, 13861425, 120157, 10.1016/j.saa.2021.120157 | |
271. | Jiali Yang, Qiqi Fu, Hui Jiang, Yinghua Li, Muqing Liu, Progress of phototherapy for osteosarcoma and application prospect of blue light photobiomodulation therapy, 2022, 12, 2234-943X, 10.3389/fonc.2022.1022973 | |
272. | Mateus Domingues Miachon, Nathali Cordeiro Pinto, Stella Regina Zamuner, Maria Cristina Chavantes, Analysis of the Potential of Blood Transvascular Sublingual with Light-Emitting Diode Irradiation in COVID-19 Patients: A Pilot Clinical Study, 2022, 40, 2578-5478, 622, 10.1089/photob.2021.0177 | |
273. | Renan Lovisetto, Tainá Caroline dos Santos Malavazzi, Lucas Andreo, Maria Fernanda Setubal Destro Rodrigues, Sandra Kalil Bussadori, Kristianne Porta Santos Fernandes, Raquel Agnelli Mesquita-Ferrari, Photobiomodulation Using Different Infrared Light Sources Promotes Muscle Precursor Cells Migration and Proliferation, 2022, 9, 2304-6732, 469, 10.3390/photonics9070469 | |
274. | Paul T. Morse, Junmei Wan, Jamie Bell, Icksoo Lee, Dennis J. Goebel, Moh H. Malek, Thomas H. Sanderson, Maik Hüttemann, Sometimes less is more: inhibitory infrared light during early reperfusion calms hyperactive mitochondria and suppresses reperfusion injury, 2022, 50, 0300-5127, 1377, 10.1042/BST20220446 | |
275. | Chung-Min Tsai, Shwu-Fen Chang, Hsi Chang, Transcranial photobiomodulation add-on therapy to valproic acid for pentylenetetrazole-induced seizures in peripubertal rats, 2022, 22, 2662-7671, 10.1186/s12906-022-03562-9 | |
276. | Juliette Hepburn, Susan Williams-Lockhart, René Jean Bensadoun, Reem Hanna, A Novel Approach of Combining Methylene Blue Photodynamic Inactivation, Photobiomodulation and Oral Ingested Methylene Blue in COVID-19 Management: A Pilot Clinical Study with 12-Month Follow-Up, 2022, 11, 2076-3921, 2211, 10.3390/antiox11112211 | |
277. | Alba Gutiérrez-Menéndez, Juan A. Martínez, Marta Méndez, Jorge L. Arias, No Effects of Photobiomodulation on Prefrontal Cortex and Hippocampal Cytochrome C Oxidase Activity and Expression of c-Fos Protein of Young Male and Female Rats, 2022, 16, 1662-453X, 10.3389/fnins.2022.897225 | |
278. | Ingvill Fjell Naterstad, Jon Joensen, Jan Magnus Bjordal, Christian Couppé, Rodrigo Alvaro Brandão Lopes-Martins, Martin Bjørn Stausholm, Efficacy of low-level laser therapy in patients with lower extremity tendinopathy or plantar fasciitis: systematic review and meta-analysis of randomised controlled trials, 2022, 12, 2044-6055, e059479, 10.1136/bmjopen-2021-059479 | |
279. | Samira Falahatdoost, Andre Chambers, Alastair Stacey, Steven Prawer, Arman Ahnood, Towards optical neuromodulation using nitrogen-doped ultrananocrystalline diamond photoelectrodes, 2022, 30, 24680230, 101850, 10.1016/j.surfin.2022.101850 | |
280. | Lucas D Dias, Kate C Blanco, Clara M G de Faria, Cristine Dozza, Elissandra M Zanchin, Fernanda R Paolillo, Kely Zampieri, Karen C Laurenti, Karina J O Souza, Juliana S A Bruno, Marcela Sene-Fiorese, Maria C C Pinto, Patrícia E Tamae, Liciane T Bello, Rosane F Z Lizarelli, Vitor H Panhoca, Antonio E de Aquino Júnior, Vanderlei S Bagnato, Perspectives on photobiomodulation and combined light-based therapies for rehabilitation of patients after COVID-19 recovery, 2022, 19, 1612-2011, 045604, 10.1088/1612-202X/ac52f5 | |
281. | Karina Alexandra Batista da Silva Freitas, Eliana Maria Minicucci, Valéria Flávia Batista da Silva, Benedito Donizete Menozzi, Hélio Langoni, Regina Célia Popim, Effects of photobiomodulation (660 nm laser) on anthracycline extravasation: An experimental study, 2022, 30, 1518-8345, 10.1590/1518-8345.5786.3693 | |
282. | Reem Hanna, Snehal Dalvi, René Jean Bensadoun, Stefano Benedicenti, Role of Photobiomodulation Therapy in Modulating Oxidative Stress in Temporomandibular Disorders. A Systematic Review and Meta-Analysis of Human Randomised Controlled Trials, 2021, 10, 2076-3921, 1028, 10.3390/antiox10071028 | |
283. | Laura Marinela Ailioaie, Constantin Ailioaie, Gerhard Litscher, Light as a Cure in COVID-19: A Challenge for Medicine, 2022, 9, 2304-6732, 686, 10.3390/photonics9100686 | |
284. | Clery Saad Abboud, Eloíza Helena da Silva Brandão, Karen Roberta Lopes Cunha, Kaline Sousa Brito, Camila de Barros Gallo, Angela Cristina Molon, Anna Carolina Ratto Tempestini Horliana, Adriana Santos Lino Franco, Kobkan Thongprasom, Maria Fernanda Setúbal Destro Rodrigues, Serum and salivary cytokines in patients with oral lichen planus treated with Photobiomodulation, 2021, 1354-523X, 10.1111/odi.14084 | |
285. | SangJoon Mo, Eun Young Kim, Yi-Suk Kwon, Min Young Lee, Jin Chul Ahn, NF-κB-mediated anti-inflammatory effects of an organic light-emitting diode (OLED) device in lipopolysaccharide (LPS)-induced in vitro and in vivo inflammation models, 2022, 13, 1664-3224, 10.3389/fimmu.2022.1050908 | |
286. | Karina Torres Pomini, Daniela Vieira Buchaim, Ana Carolina Cestari Bighetti, Abdul Latif Hamzé, Carlos Henrique Bertoni Reis, Marco Antonio Húngaro Duarte, Murilo Priori Alcalde, Benedito Barraviera, Rui Seabra Ferreira Júnior, Alexandre Teixeira de Souza, Paulo Sérgio da Silva Santos, João Paulo Galletti Pilon, Miguel Ângelo de Marchi, Dayane Maria Braz Nogueira, Cleuber Rodrigo de Souza Bueno, Wendel Cleber Soares, Rogerio Leone Buchaim, Tissue Bioengineering with Fibrin Scaffolds and Deproteinized Bone Matrix Associated or Not with the Transoperative Laser Photobiomodulation Protocol, 2023, 28, 1420-3049, 407, 10.3390/molecules28010407 | |
287. | Belkiss Câmara Mármora, Fernanda Thomé Brochado, Tuany Rafaelli Schmidt, Lucas Gonçalves Santos, Aurigena Antunes de Araújo, Caroline Addison Carvalho Xavier de Medeiros, Susana Barbosa Ribeiro, Marco Antonio Trevizani Martins, Emily Ferreira Salles Pilar, Vivian Petersen Wagner, Manoela Domingues Martins, Defocused high-power diode laser accelerates skin repair in a murine model through REDOX state modulation and reepithelization and collagen deposition stimulation, 2021, 225, 10111344, 112332, 10.1016/j.jphotobiol.2021.112332 | |
288. | Hannah Serrage, Vladimir Heiskanen, William M. Palin, Paul R. Cooper, Michael R. Milward, Mohammed Hadis, Michael R. Hamblin, Under the spotlight: mechanisms of photobiomodulation concentrating on blue and green light, 2019, 18, 1474-905X, 1877, 10.1039/c9pp00089e | |
289. | Patricia Kasowanjete, Nicolette N Houreld, Heidi Abrahamse, The effect of photomodulation on fibroblast growth factor and the Ras/MAPK signalling pathway: a review, 2022, 31, 0969-0700, 832, 10.12968/jowc.2022.31.10.832 | |
290. | Letícia Mello Bezinelli, Luciana Corrêa, Cristina Vogel, Jose Mauro Kutner, Andreza Feitosa Ribeiro, Nelson Hamerschlak, Carlos de Paula Eduardo, Cesar Augusto Migliorati, Fernanda de Paula Eduardo, Long-term safety of photobiomodulation therapy for oral mucositis in hematopoietic cell transplantation patients: a 15-year retrospective study, 2021, 29, 0941-4355, 6891, 10.1007/s00520-021-06268-9 | |
291. | Naresh Kumar Rajendran, Nicolette Nadene Houreld, Photobiomodulation hastens diabetic wound healing via modulation of the PI3K/AKT/FoxO1 pathway in an adipose derived stem cell-fibroblast co-culture, 2022, 12, 26664690, 100157, 10.1016/j.jpap.2022.100157 | |
292. | Pâmela Camila Pereira, Carlos José de Lima, Adriana Barrinha Fernandes, Fernanda Barrinha Fernandes, Renato Amaro Zângaro, Antonio Balbin Villaverde, Systemic Effects of Photobiomodulation on Blood Components in the Treatment of Community-Acquired Pneumonia, 2022, 40, 2578-5478, 51, 10.1089/photob.2021.0050 | |
293. | Lidong Huang, Weiyu Gong, Guibin Huang, Jingyi Li, Jilin Wu, Yuguang Wang, Yanmei Dong, The additive effects of photobiomodulation and bioactive glasses on enhancing early angiogenesis, 2022, 17, 1748-6041, 045007, 10.1088/1748-605X/ac6b07 | |
294. | Asma Shaikh-Kader, Nicolette Nadene Houreld, Photobiomodulation, Cells of Connective Tissue and Repair Processes: A Look at In Vivo and In Vitro Studies on Bone, Cartilage and Tendon Cells, 2022, 9, 2304-6732, 618, 10.3390/photonics9090618 | |
295. | M Patmadevi, A T Mukti, A S Mubarak, S D Astuti, The Effect of Laserpuncture on Accelerate Gonadal Maturity of Female Striped Catfish (Pangasianodon hypophthalmus), 2022, 1036, 1755-1307, 012036, 10.1088/1755-1315/1036/1/012036 | |
296. | Stephanie Maxine Ross, Integrative Pain Solutions, Part 3, 2022, 36, 0887-9311, 327, 10.1097/HNP.0000000000000549 | |
297. | Clara Maria Gonçalves de Faria, Heloisa Ciol, Vanderlei Salvador Bagnato, Sebastião Pratavieira, Effects of photobiomodulation on the redox state of healthy and cancer cells, 2021, 12, 2156-7085, 3902, 10.1364/BOE.421302 | |
298. | Hyunha Kim, Min Jae Kim, Young Woo Kwon, Sangheon Jeon, Seo‐Yeon Lee, Chang‐Seok Kim, Byung Tae Choi, Yong‐Il Shin, Suck Won Hong, Hwa Kyoung Shin, Benefits of a Skull‐Interfaced Flexible and Implantable Multilight Emitting Diode Array for Photobiomodulation in Ischemic Stroke, 2022, 9, 2198-3844, 2104629, 10.1002/advs.202104629 | |
299. | Francesco Inchingolo, Denisa Hazballa, Alessio Danilo Inchingolo, Giuseppina Malcangi, Grazia Marinelli, Antonio Mancini, Maria Elena Maggiore, Ioana Roxana Bordea, Antonio Scarano, Marco Farronato, Gianluca Martino Tartaglia, Felice Lorusso, Angelo Michele Inchingolo, Gianna Dipalma, Innovative Concepts and Recent Breakthrough for Engineered Graft and Constructs for Bone Regeneration: A Literature Systematic Review, 2022, 15, 1996-1944, 1120, 10.3390/ma15031120 | |
300. | Qiannan Liu, Hao Wu, Hanxu Zhang, Yue Pan, Siqi Du, Wuqi Song, Fengmin Zhang, Hailiang Liu, Heat Shock Protein Is Associated with Inhibition of Inflammatory Cytokine Production by 630 nm Light-Emitting Diode Irradiation in Fibroblast-Like Synoviocytes Based on RNA Sequencing Analysis, 2022, 40, 2578-5478, 751, 10.1089/photob.2022.0041 | |
301. | Brian Bicknell, E-Liisa Laakso, Ann Liebert, Hosen Kiat, Modifying the Microbiome as a Potential Mechanism of Photobiomodulation: A Case Report, 2022, 40, 2578-5478, 88, 10.1089/photob.2021.0057 | |
302. | Amir Hossein Ghaderi, Ali Jahan, Fatemeh Akrami, Maryam Moghadam Salimi, Transcranial photobiomodulation changes topology, synchronizability, and complexity of resting state brain networks, 2021, 18, 1741-2560, 046048, 10.1088/1741-2552/abf97c | |
303. | Piotr Rola, Szymon Włodarczak, Maciej Lesiak, Adrian Doroszko, Adrian Włodarczak, Changes in Cell Biology under the Influence of Low-Level Laser Therapy, 2022, 9, 2304-6732, 502, 10.3390/photonics9070502 | |
304. | Ann Liebert, Brian Bicknell, E-Liisa Laakso, Gillian Heller, Parastoo Jalilitabaei, Sharon Tilley, John Mitrofanis, Hosen Kiat, Improvements in clinical signs of Parkinson’s disease using photobiomodulation: a prospective proof-of-concept study, 2021, 21, 1471-2377, 10.1186/s12883-021-02248-y | |
305. | Frederico Carlos Jana Neto, Ana Luiza Cabrera Martimbianco, Raquel Agnelli Mesquita‐Ferrari, Sandra Kalil Bussadori, Gustavo Porto Alves, Paulo Victor Dias Almeida, Felipe Guimaraes Delgado, Lucas Resende Fonseca, Maiara Zeni Graiff Gama, Mariana Djrdjrjan Jorge, Michael R. Hamblin, Kristianne Porta Santos Fernandes, Effects of multiwavelength photobiomodulation for the treatment of traumatic soft tissue injuries associated with bone fractures: A double‐blind, randomized controlled clinical trial, 2023, 1864-063X, 10.1002/jbio.202200299 | |
306. | Faezeh Fallahi, Atarodalsadat Mostafavinia, Zahranadia sharifi, Leila Mohaghegh Shalmani, Abdollah Amini, Houssein Ahmadi, Hamidreza Omidi, Masoumeh Hajihosseintehrani, Sahar Bayat, Michael R. Hamblin, Sufan Chien, Mohammad Bayat, Effects of photobiomodulation on mitochondrial function in diabetic adipose-derived stem cells in vitro, 2023, 285, 13861425, 121835, 10.1016/j.saa.2022.121835 | |
307. | Fábio Pericinoto Giolo, Gabriel Silva Santos, Victor Fontes Pacheco, Stephany Cares Huber, Kaue Franco Malange, Bruno Lima Rodrigues, Fernanda Bassora, Tomas Mosaner, Gabriel Azzini, Lucas Leite Ribeiro, Carlos Amilcar Parada, José Fábio Santos Duarte Lana, Photobiomodulation therapy for osteoarthritis: Mechanisms of action, 2022, 10, 2220-6132, 29, 10.5528/wjtm.v10.i3.29 | |
308. | Gisela C. V. Camolesi, José M. Somoza‐Martín, Maria D. Reboiras‐López, Fabio Camacho‐Alonso, Andrés Blanco‐Carrión, Mario Pérez‐Sayáns, Photobiomodulation in dental implant stability and post‐surgical healing and inflammation. A randomised double‐blind study, 2023, 34, 0905-7161, 137, 10.1111/clr.14026 | |
309. | Armin Khosravipour, Abdollah Amini, Reza Masteri Farahani, Atarodalsadat Mostafavinia, Mehrdad Asgari, Fatemehalsadat Rezaei, Heidi Abrahamse, Sufan Chien, Mohammad Bayat, Evaluation of the effects of preconditioned human stem cells plus a scaffold and photobiomodulation administration on stereological parameters and gene expression levels in a critical size bone defect in rats, 2022, 37, 1435-604X, 2457, 10.1007/s10103-022-03509-z | |
310. | Jade I Basem, Tiffany Lin, Neel D Mehta, A Comprehensive Review: Chronic Pain Sequelae in the Presence of Ehlers–Danlos Syndrome, 2022, 26, 1531-3433, 871, 10.1007/s11916-022-01093-z | |
311. | Guilherme Aparecido Monteiro Duque da Fonseca, Marcos Fernando Xisto Braga Cavalcanti, José Daniel de Souza Maior, Juliana da Silva Pereira, Leandro augusto Pinto, Murilo Matias, Lucio Frigo, Laser-photobiomodulation on titanium implant bone healing in rat model: comparison between 660- and 808-nm wavelength, 2022, 37, 1435-604X, 2179, 10.1007/s10103-021-03481-0 | |
312. | Camila Amaral Coracini, Matheus Felipe Zazula, Mariane Okamoto Ferreira, Janaina Carla da Silva, Suellen Ribeiro da Silva Scarton, Carolina Panis, Lucinéia de Fátima Chasko Ribeiro, Taciane Stein da Silva Leal, Gladson Ricardo Flor Bertolini, Acute effects of photobiomodulation applied on the dorsal root ganglion in gout model-induced rats, 2023, 239, 10111344, 112644, 10.1016/j.jphotobiol.2022.112644 | |
313. | Karina Alexandra Batista da Silva Freitas, Eliana Maria Minicucci, Valéria Flávia Batista da Silva, Benedito Donizete Menozzi, Hélio Langoni, Regina Célia Popim, Efectos de la fotobiomodulación (láser de 660 nm) sobre la extravasación de antraciclinas: estudio experimental, 2022, 30, 1518-8345, 10.1590/1518-8345.5786.3692 | |
314. | Piotr Tkocz, Tomasz Matusz, Łukasz Kosowski, Karolina Walewicz, Łukasz Argier, Michał Kuszewski, Magdalena Hagner-Derengowska, Kuba Ptaszkowski, Robert Dymarek, Jakub Taradaj, A Randomised-Controlled Clinical Study Examining the Effect of High-Intensity Laser Therapy (HILT) on the Management of Painful Calcaneal Spur with Plantar Fasciitis, 2021, 10, 2077-0383, 4891, 10.3390/jcm10214891 | |
315. | Philip M. Preshaw, Mark Ide, Susan M. Bissett, Richard Holliday, Nichola Lansdowne, Kimberley Pickering, Judy A. Taylor, Ana M. Levonian, Christine Pleasance, Maria Elena Guarnelli, Anna Simonelli, Chiara Fabbri, Roberto Farina, Fotinos S. Panagakos, Leonardo Trombelli, No benefit of an adjunctive phototherapy protocol in treatment of periodontitis: A split‐mouth randomized controlled trial, 2021, 48, 0303-6979, 1093, 10.1111/jcpe.13465 | |
316. | Polina Bikmulina, Nastasia Kosheleva, Anastasia Shpichka, Vladimir Yusupov, Vladimir Gogvadze, Yury Rochev, Peter Timashev, Photobiomodulation in 3D tissue engineering, 2022, 27, 1083-3668, 10.1117/1.JBO.27.9.090901 | |
317. | Yurii V. Stepanov, Iuliia Golovynska, Sergii Golovynskyi, Liudmyla V. Garmanchuk, Oleksandr Gorbach, Liudmyla I. Stepanova, Natalia Khranovska, Liudmyla I. Ostapchenko, Tymish Y. Ohulchanskyy, Junle Qu, Red and near infrared light-stimulated angiogenesis mediated via Ca2+ influx, VEGF production and NO synthesis in endothelial cells in macrophage or malignant environments, 2022, 227, 10111344, 112388, 10.1016/j.jphotobiol.2022.112388 | |
318. | Hyun Seok Ryu, Nam Kyu Lim, Andrew Reyes Padalhin, Celine Abueva, So Young Park, Phil‐sang Chung, Seung Hoon Woo, Improved healing and macrophage polarization in oral ulcers treated with photobiomodulation (PBM), 2022, 54, 0196-8092, 600, 10.1002/lsm.23510 | |
319. | Pâmela Camila Pereira, Carlos José de Lima, Adriana Barrinha Fernandes, Renato Amaro Zângaro, Antonio Balbin Villaverde, Cardiopulmonary and hematological effects of infrared LED photobiomodulation in the treatment of SARS-COV2, 2023, 238, 10111344, 112619, 10.1016/j.jphotobiol.2022.112619 | |
320. | Fernanda Thomé Brochado, Belkiss Câmara Mármora, Paloma Santos Campos, Tuany Rafaeli Schmidt, Kristianne Porta Santos Fernandes, Sandra Kalil Bussadori, Lucas Gonçalves Santos, Vivian Petersen Wagner, Marcelo Lazzaron Lamers, Manoela Domingues Martins, Effects of different protocols of defocused high-power laser on the viability and migration of myoblasts—a comparative in vitro study, 2022, 37, 1435-604X, 3571, 10.1007/s10103-022-03636-7 | |
321. | Mojgan Shavakhi, Shabnam Tahamtan, Maryam Saki, Reza Fekrazad, Effectiveness of Photobiomodulation on Orthodontic Mini Screw Stability: A Systematic Review, 2021, 39, 2578-5478, 747, 10.1089/photob.2021.0081 | |
322. | Nathali Cordeiro Pinto, Marcelo Victor Pires de, Nathalia Lopes Ferreira, Natalia Almeida Braga, Alexandre Aldred, Guilherme Gomes, George Miguel Góes Freire, Hazem Adel Ashmawi, Marucia Chacur, Customized Photobiomodulation Modulates Pain and Alters Thermography Pattern in Patients with Knee Osteoarthritis: A Randomized Double-Blind Pilot Study, 2022, 40, 2578-5478, 698, 10.1089/photob.2022.0067 | |
323. | Peiyu Jiang, Yunyi Liu, Jiawen Zhang, Yixuan Liu, Min Li, Meng Tao, Yue Zhang, Zongxiang Tang, Wentao Liu, Yang Xu, Mast cell stabilization: new mechanism underlying the therapeutic effect of intense pulsed light on rosacea, 2023, 72, 1023-3830, 75, 10.1007/s00011-022-01635-6 | |
324. | Margaret A. Naeser, Paula I. Martin, Michael D. Ho, Maxine H. Krengel, Yelena Bogdanova, Jeffrey A. Knight, Michael R. Hamblin, Andrea E. Fedoruk, Luke G. Poole, ChiaHsin Cheng, BangBon Koo, Transcranial Photobiomodulation Treatment: Significant Improvements in Four Ex-Football Players with Possible Chronic Traumatic Encephalopathy, 2023, 7, 25424823, 77, 10.3233/ADR-220022 | |
325. | Samantha Ketelyn Silva, Ana Maria Guzzi Plepis, Virginia da Conceição Amaro Martins, Marilia Marta Horn, Daniela Vieira Buchaim, Rogerio Leone Buchaim, André Antônio Pelegrine, Vinícius Rodrigues Silva, Mateus Hissashi Matsumoto Kudo, José Francisco Rebello Fernandes, Fabricio Montenegro Nazari, Marcelo Rodrigues da Cunha, Suitability of Chitosan Scaffolds with Carbon Nanotubes for Bone Defects Treated with Photobiomodulation, 2022, 23, 1422-0067, 6503, 10.3390/ijms23126503 | |
326. | Soofia Naghdi, Noureddin Nakhostin Ansari, Mitra Varedi, Mojtaba Fathali, Milad Zarrin, Mina Kashi-Alashti, Fatemeh HasanNia, Use of low-level laser therapy for patients with chronic rhinosinusitis: a single-blind, sham-controlled clinical trial, 2022, 38, 1435-604X, 10.1007/s10103-022-03684-z | |
327. | Thamyres Maria Silva Simões, José de Alencar Fernandes Neto, Cassiano Francisco Weege Nonaka, Maria Helena Chaves de Vasconcelos Catão, Effects of photobiomodulation therapy with red LED on inflammatory cells during the healing of skin burns, 2022, 37, 1435-604X, 2817, 10.1007/s10103-022-03537-9 | |
328. | Ann Liebert, Brian Bicknell, Wayne Markman, Hosen Kiat, A Potential Role for Photobiomodulation Therapy in Disease Treatment and Prevention in the Era of COVID-19, 2020, 11, 2152-5250, 1352, 10.14336/AD.2020.0901 | |
329. | Karina Torres Pomini, Daniela Vieira Buchaim, Ana Carolina Cestari Bighetti, Jesus Carlos Andreo, Marcelie Priscila de Oliveira Rosso, José Stalin Bayas Escudero, Bruna Botteon Della Coletta, Murilo Priori Alcalde, Marco Antonio Hungaro Duarte, Dimitrius Leonardo Pitol, João Paulo Mardegan Issa, Edilson Ervolino, Matheus Bento Medeiros Moscatel, Márcia Zilioli Bellini, Alexandre Teixeira de Souza, Wendel Cleber Soares, Rogerio Leone Buchaim, Use of Photobiomodulation Combined with Fibrin Sealant and Bone Substitute Improving the Bone Repair of Critical Defects, 2022, 14, 2073-4360, 4170, 10.3390/polym14194170 | |
330. | Ievgeniia Kocherova, Artur Bryja, Katarzyna Błochowiak, Mariusz Kaczmarek, Katarzyna Stefańska, Jacek Matys, Kinga Grzech-Leśniak, Marzena Dominiak, Paul Mozdziak, Bartosz Kempisty, Marta Dyszkiewicz-Konwińska, Photobiomodulation with Red and Near-Infrared Light Improves Viability and Modulates Expression of Mesenchymal and Apoptotic-Related Markers in Human Gingival Fibroblasts, 2021, 14, 1996-1944, 3427, 10.3390/ma14123427 | |
331. | O. V. Ketsa, M. O. Zelinska, M. M. Marchenko, Proteins oxidative modification and antioxidant enzymes activity in the liver mitochondria of rats under laser irradiation and administration of ω-3 polyunsaturated fatty acids, 2022, 94, 24094943, 44, 10.15407/ubj94.01.044 | |
332. | Andrew McColloch, Hanli Liu, Michael Cho, Reversal of stem cell‐derived hypertrophic adipocytes mediated by photobiomodulation (1064 nm), 2021, 3, 2627-1850, 10.1002/tbio.202100006 | |
333. | Salvatore Saieva, Giulio Taglialatela, Near-infrared light reduces glia activation and modulates neuroinflammation in the brains of diet-induced obese mice, 2022, 12, 2045-2322, 10.1038/s41598-022-14812-8 | |
334. | Marc Argilés, Bernat Sunyer-Grau, Sílvia Arteche-Fernandez, Cleofé Peña-Gómez, Functional connectivity of brain networks with three monochromatic wavelengths: a pilot study using resting-state functional magnetic resonance imaging, 2022, 12, 2045-2322, 10.1038/s41598-022-20668-9 | |
335. | Xinlu Gao, Wenwen Zhang, Fan Yang, Wenya Ma, Benzhi Cai, Andrey J. Serra, Photobiomodulation Regulation as One Promising Therapeutic Approach for Myocardial Infarction, 2021, 2021, 1942-0994, 1, 10.1155/2021/9962922 | |
336. | Sándor Sandra, A COVID-19 (koronavírus okozta megbetegedés) kezelési lehetőségei a fizioterápia eszközeivel, kiemelt figyelemmel a fototerápia eddigi eredményeire, 2021, 69, 2677-1632, 431, 10.38146/BSZ.2021.3.5 | |
337. | Miguel Tolentino, Chi C. Cho, Jeri-Anne Lyons, Photobiomodulation Modulates Interleukin-10 and Interferon Gamma Production by Mononuclear Cells from Healthy Donors and Persons with Multiple Sclerosis, 2022, 40, 2578-5478, 234, 10.1089/photob.2021.0169 | |
338. | Marco H. Ji, Alexander Kreymerman, Kinsley Belle, Benjamin K. Ghiam, Stephanie P. Muscat, Vinit B. Mahajan, Gregory M. Enns, Mark Mercola, Edward H. Wood, The Present and Future of Mitochondrial-Based Therapeutics for Eye Disease, 2021, 10, 2164-2591, 4, 10.1167/tvst.10.8.4 | |
339. | Damir Nizamutdinov, Xiaoming Qi, Marvin H Berman, Gordon Dougal, Samantha Dayawansa, Erxi Wu, S. Stephen Yi, Alan B Stevens, Jason H Huang, Transcranial Near Infrared Light Stimulations Improve Cognition in Patients with Dementia, 2021, 12, 2152-5250, 954, 10.14336/AD.2021.0229 | |
340. | Natalia Osorio Viarengo, Marcela Leticia Leal Gonçalves, Laura Hermida Bruno, Ana Laura Fossati, María Roxana Ferreira Sertaje, Elaine Marcilio Santos, Ana Paula Taboada Sobral, Raquel Agnelli Mesquita-Ferrari, Kristianne Porta Santos Fernandes, Anna Carolina Ratto Tempestini Horliana, Lara Jansiski Motta, Sandra Kalil Bussadori, Evaluation of intravascular irradiation of blood in children with sleep bruxism: Study protocol for a randomized controlled clinical trial, 2022, 101, 1536-5964, e31230, 10.1097/MD.0000000000031230 | |
341. | Yago M. Dutra, Elvis S. Malta, Amanda S. Elias, James R. Broatch, Alessandro M. Zagatto, Deconstructing the Ergogenic Effects of Photobiomodulation: A Systematic Review and Meta-analysis of its Efficacy in Improving Mode-Specific Exercise Performance in Humans, 2022, 52, 0112-1642, 2733, 10.1007/s40279-022-01714-y | |
342. | L. G. C. Corrêa, D. S. F. Magalhães, A. Baptista, A. F. Frade-Barros, 2022, Chapter 181, 978-3-030-70600-5, 1211, 10.1007/978-3-030-70601-2_181 | |
343. | Neeraj Kohli, Barry Jarnagin, Angela R Stoehr, Georgine Lamvu, An observational cohort study of pelvic floor photobiomodulation for treatment of chronic pelvic pain, 2021, 10, 2042-6305, 1291, 10.2217/cer-2021-0187 | |
344. | Karina Alexandra Batista da Silva Freitas, Eliana Maria Minicucci, Valéria Flávia Batista da Silva, Benedito Donizete Menozzi, Hélio Langoni, Regina Célia Popim, Efeitos da fotobiomodulação (laser 660 nm) no extravasamento de antraciclina: estudo experimental, 2022, 30, 1518-8345, 10.1590/1518-8345.5786.3694 | |
345. | Zeqing Chen, Shijie Huang, Muqing Liu, The review of the light parameters and mechanisms of Photobiomodulation on melanoma cells, 2022, 38, 0905-4383, 3, 10.1111/phpp.12715 | |
346. | Shinya Yokomizo, Wataru Katagiri, Yohei Maki, Tomoya Sano, Kazumasa Inoue, Masahiro Fukushi, Dmitriy N. Atochin, Toshihiro Kushibiki, Akihiko Kawana, Yoshifumi Kimizuka, Satoshi Kashiwagi, Brief exposure of skin to near-infrared laser augments early vaccine responses, 2021, 10, 2192-8614, 3187, 10.1515/nanoph-2021-0133 | |
347. | Shuang-Qiao Sun, Jing-Jing Shen, Yu-Fei Wang, Yu-Tong Jiang, Lin-Fu Chen, Hua Xin, Jiang-Nan Wang, Xiao-Bo Shi, Xiao-Zhao Zhu, Qi Sun, Liang-Sheng Liao, Qian Chen, Man-Keung Fung, Shuit-Tong Lee, Red organic light-emitting diodes based photobiomodulation therapy enabling prominent hair growth, 2023, 1998-0124, 10.1007/s12274-022-5315-1 | |
348. | Sandra R.C.A. Fagnani, Susana C.P.S. de Oliveira, Juliana S.C. Monteiro, Fernando José Pires Sampaio, Pedro J.L. Crugeira, Jean N. dos Santos, Antonio L.B. Pinheiro, Histological evaluation of skin lesions induced by Leishmania braziliensis treated by PACT using Laser light and 1.9 dimethyl-methylene blue, 2022, 38, 15721000, 102815, 10.1016/j.pdpdt.2022.102815 | |
349. | Ana Carolina Lemos Pimentel, Tânia Tavares Rodriguez, Manoela Domingues Martins, Luiz Carlos Ramos Filho, Iago Fonseca Silva Mota, Juliana Santos de Carvalho Monteiro, Patricia Rubia Manieri, Antônio Luiz Barbosa Pinheiro, Patrícia Ramos Cury, Jean Nunes Dos Santos, Impact of photobiomodulation therapy on the morphological aspects of submandibular gland submitted to excretory duct ligation and hypothyroidism: an animal study, 2022, 37, 1435-604X, 2005, 10.1007/s10103-021-03463-2 | |
350. | Katayoon Montazeri, Mohammad Farhadi, Reza Fekrazad, Samira Chaibakhsh, Saeid Mahmoudian, Photobiomodulation therapy in mood disorders: a systematic review, 2022, 37, 1435-604X, 3343, 10.1007/s10103-022-03641-w | |
351. | Pooya Ebrahimi, Mahdi Hadilou, Ferdos Naserneysari, Amirmohammad Dolatabadi, Rana Tarzemany, Nafiseh Vahed, Leila Nikniaz, Reza Fekrazad, Leila Gholami, Effect of photobiomodulation in secondary intention gingival wound healing—a systematic review and meta-analysis, 2021, 21, 1472-6831, 10.1186/s12903-021-01611-2 | |
352. | Sulbha K. Sharma, Sakshi Sardana, Michael R. Hamblin, Role of opsins and light or heat activated transient receptor potential ion channels in the mechanisms of photobiomodulation and infrared therapy, 2023, 13, 26664690, 100160, 10.1016/j.jpap.2023.100160 | |
353. | Mirele Regina Silva, Amanda Rodrigues Scheffer, Roberto Sávio de Assunção Bastos, Maria Cristina Chavantes, Maria Fernanda Capoani Garcia Mondelli, The effects of photobiomodulation therapy in individuals with tinnitus and without hearing loss, 2022, 37, 1435-604X, 3485, 10.1007/s10103-022-03614-z | |
354. | Saeed Vafaei-Nezhad, Somayeh Niknazar, Ali Asghar Payvandi, Atefeh Shirazi Tehrani, Navid Ahmady Roozbahany, Behnaz Ahrabi, Hojjat Allah Abbaszadeh, Shahram Darabi, Therapeutic Effects of Photobiomodulation Therapy on Multiple Sclerosis by Regulating the Inflammatory Process and Controlling Immune Cell Activity: A Novel Promising Treatment Target, 2022, 13, 2008-9783, e32, 10.34172/jlms.2022.32 | |
355. | Alistair V. W. Nunn, Geoffrey W. Guy, Wolfgang Brysch, Jimmy D. Bell, Understanding Long COVID; Mitochondrial Health and Adaptation—Old Pathways, New Problems, 2022, 10, 2227-9059, 3113, 10.3390/biomedicines10123113 | |
356. | Lauren E. Rentz, Randy W. Bryner, Jad Ramadan, Ali Rezai, Scott M. Galster, Full-Body Photobiomodulation Therapy Is Associated with Reduced Sleep Durations and Augmented Cardiorespiratory Indicators of Recovery, 2022, 10, 2075-4663, 119, 10.3390/sports10080119 | |
357. | Erick Frank Bragato, Jefferson André Pires, Marcos Momolli, Marina Bertoni Guerra, Adriana Fernandes Paisano, Raquel Agnelli Mesquita Ferrari, Sandra Kalil Bussadori, Lara Jansiski Motta, Kristianne Porta Santos Fernandes, Comparison of the effects of 2 frequencies of application of photobiomodulation on facial rejuvenation: Controlled, randomized, and double-blind clinical trial, 2023, 102, 0025-7974, e32514, 10.1097/MD.0000000000032514 | |
358. | Namgue Hong, Hee Jung Kim, Keunsoo Kang, Ji On Park, Seyoung Mun, Hyung-Gun Kim, Bong Hui Kang, Phil-Sang Chung, Min Young Lee, Jin-Chul Ahn, Photobiomodulation improves the synapses and cognitive function and ameliorates epileptic seizure by inhibiting downregulation of Nlgn3, 2023, 13, 2045-3701, 10.1186/s13578-022-00949-6 | |
359. | Elisabetta Gerace, Francesca Cialdai, Elettra Sereni, Daniele Lana, Daniele Nosi, Maria Grazia Giovannini, Monica Monici, Guido Mannaioni, NIR Laser Photobiomodulation Induces Neuroprotection in an In Vitro Model of Cerebral Hypoxia/Ischemia, 2021, 58, 0893-7648, 5383, 10.1007/s12035-021-02496-6 | |
360. | Blanche Aguida, Marootpong Pooam, Margaret Ahmad, Nathalie Jourdan, Infrared light therapy relieves TLR-4 dependent hyper-inflammation of the type induced by COVID-19, 2021, 14, 1942-0889, 200, 10.1080/19420889.2021.1965718 | |
361. | Tyrell Pruitt, Caroline Carter, Xinlong Wang, Anqi Wu, Hanli Liu, Photobiomodulation at Different Wavelengths Boosts Mitochondrial Redox Metabolism and Hemoglobin Oxygenation: Lasers vs. Light-Emitting Diodes In Vivo, 2022, 12, 2218-1989, 103, 10.3390/metabo12020103 | |
362. | Xiaozhong Wu, Qinglei Guo, Bioresorbable Photonics: Materials, Devices and Applications, 2021, 8, 2304-6732, 235, 10.3390/photonics8070235 | |
363. | Chun-Xia Zhang, Yan Lou, Jing Chi, Xiao-Li Bao, Bin Fan, Guang-Yu Li, Considerations for the Use of Photobiomodulation in the Treatment of Retinal Diseases, 2022, 12, 2218-273X, 1811, 10.3390/biom12121811 | |
364. | Narda G. Robinson, 2022, 9781119791195, 330, 10.1002/9781119791256.ch16 | |
365. | Fabrízio dos Santos Cardoso, Farzad Salehpour, Norberto Cysne Coimbra, Francisco Gonzalez-Lima, Sérgio Gomes da Silva, Photobiomodulation for the treatment of neuroinflammation: A systematic review of controlled laboratory animal studies, 2022, 16, 1662-453X, 10.3389/fnins.2022.1006031 | |
366. | Ji Bian, Ann Liebert, Brian Bicknell, Xin-Ming Chen, Chunling Huang, Carol A. Pollock, Therapeutic Potential of Photobiomodulation for Chronic Kidney Disease, 2022, 23, 1422-0067, 8043, 10.3390/ijms23148043 | |
367. | Priyanka Shaw, Naresh Kumar, Maxime Sahun, Evelien Smits, Annemie Bogaerts, Angela Privat-Maldonado, Modulating the Antioxidant Response for Better Oxidative Stress-Inducing Therapies: How to Take Advantage of Two Sides of the Same Medal?, 2022, 10, 2227-9059, 823, 10.3390/biomedicines10040823 | |
368. | Ann Liebert, Brian Bicknell, E-Liisa Laakso, Parastoo Jalilitabaei, Sharon Tilley, Hosen Kiat, John Mitrofanis, Remote Photobiomodulation Treatment for the Clinical Signs of Parkinson's Disease: A Case Series Conducted During COVID-19, 2022, 40, 2578-5478, 112, 10.1089/photob.2021.0056 | |
369. | L.M.B. Cabreira, J.K. Merlo, J.L. Jacinto, J.P. Nunes, A.S. Ribeiro, A.F. Aguiar, Photobiomodulation therapy with light-emitting diode does not improve lower-body muscle performance and delayed-onset muscle soreness in resistance-trained women: A randomized, controlled, crossover trial, 2022, 37, 07651597, 635.e1, 10.1016/j.scispo.2021.06.006 | |
370. | Ali Neshasteh-Riz, Fatemeh Ramezani, Keihan Kookli, Seyedalireza Moghaddas Fazeli, Ali Motamed, Farinaaz Nasirinezhad, Atousa Janzadeh, Michael R. Hamblin, Mohammadreza Asadi, Optimization of the Duration and Dose of Photobiomodulation Therapy (660 nm Laser) for Spinal Cord Injury in Rats, 2022, 40, 2578-5478, 488, 10.1089/photob.2022.0012 | |
371. | Ann Liebert, Neda Seyedsadjadi, Vincent Pang, Gerhard Litscher, Hosen Kiat, Evaluation of Gender Differences in Response to Photobiomodulation Therapy, Including Laser Acupuncture: A Narrative Review and Implication to Precision Medicine, 2022, 40, 2578-5478, 78, 10.1089/photob.2021.0066 | |
372. | Laura Marinela Ailioaie, Gerhard Litscher, Photobiomodulation and Sports: Results of a Narrative Review, 2021, 11, 2075-1729, 1339, 10.3390/life11121339 | |
373. | Yurii V. Stepanov, Iuliia Golovynska, Renlong Zhang, Sergii Golovynskyi, Liudmyla I. Stepanova, Oleksandr Gorbach, Taisa Dovbynchuk, Liudmyla V. Garmanchuk, Tymish Y. Ohulchanskyy, Junle Qu, Near-infrared light reduces β-amyloid-stimulated microglial toxicity and enhances survival of neurons: mechanisms of light therapy for Alzheimer’s disease, 2022, 14, 1758-9193, 10.1186/s13195-022-01022-7 | |
374. | Patricia Gabrielli Vassão, Ana Carolina Flygare de Souza, Raquel Munhoz da Silveira Campos, Livia Assis Garcia, Helga Tatiana Tucci, Ana Claudia Muniz Renno, Effects of photobiomodulation and a physical exercise program on the expression of inflammatory and cartilage degradation biomarkers and functional capacity in women with knee osteoarthritis: a randomized blinded study, 2021, 61, 2523-3106, 10.1186/s42358-021-00220-5 | |
375. | Mara Lúcia Gonçalves Diogo, Thalita Molinos Campos, Elsa Susana Reis Fonseca, Christiane Pavani, Anna Carolina Ratto Tempestini Horliana, Kristianne Porta Santos Fernandes, Sandra Kalil Bussadori, Francisca Goreth Malheiro Moraes Fantin, Diego Portes Vieira Leite, Ângela Toshie Araki Yamamoto, Ricardo Scarparo Navarro, Lara Jansiski Motta, Effect of Blue Light on Acne Vulgaris: A Systematic Review, 2021, 21, 1424-8220, 6943, 10.3390/s21206943 | |
376. | Paulo Goberlânio de Barros Silva, Edson Luiz Cetira Filho, Flávia Maria Noronha Nigri, Thinali Sousa Dantas, George Táccio de Miranda Candeiro, Jiovanne Rabelo Neri, Photobiomodulation Reduces Pain-Related Symptoms Without Interfering in the Efficacy of In-Office Tooth Bleaching: A Systematic Review and Meta-Analysis of Placebo-Controlled Clinical Trials, 2022, 40, 2578-5478, 163, 10.1089/photob.2021.0105 | |
377. | Nancy Zeaiter, Kinga Grzech-Leśniak, Zuzanna Grzech-Leśniak, Maher Ghandour, Marwan El Mobadder, Facial Aesthetic Laser-Assisted Protocol for the Management of Acne and Pigmentation: A Case Report, 2022, 2168-8184, 10.7759/cureus.28871 | |
378. | Mateus B. Souza, Rodrigo O. Mascarenhas, Laisa B. Maia, Letícia S. Fonseca, Hytalo J. Silva, Rutger M. J. de Zoete, James H. McAuley, Nicholas Henschke, Vinicius C. Oliveira, Essam Al-Moraissi, Comparative efficacy and acceptability of non-pharmacological interventions in fibromyalgia: Protocol for a network meta-analysis, 2022, 17, 1932-6203, e0274406, 10.1371/journal.pone.0274406 | |
379. | Dun-Xian Tan, Russel J. Reiter, Scott Zimmerman, Ruediger Hardeland, Melatonin: Both a Messenger of Darkness and a Participant in the Cellular Actions of Non-Visible Solar Radiation of Near Infrared Light, 2023, 12, 2079-7737, 89, 10.3390/biology12010089 | |
380. | Susana Padoin, Aline C. Zeffa, Julio C. Molina Corrêa, Tarlyson R. de Angelis, Thuany B. Moreira, Lilian K. Barazetti, Solange de Paula Ramos, Phototherapy Improves Muscle Recovery and Does Not Impair Repeated Bout Effect in Plyometric Exercise, 2022, 36, 1064-8011, 3301, 10.1519/JSC.0000000000003895 | |
381. | Zhiyi Shan, Juanjuan Ji, Colman McGrath, Min Gu, Yanqi Yang, Effects of low-level light therapy on dentin hypersensitivity: a systematic review and meta-analysis, 2021, 25, 1432-6981, 6571, 10.1007/s00784-021-04183-1 | |
382. | Roya Derakhshan, Houssein Ahmadi, Mohammad Bayat, Leila Mehboudi, Erfan Pourhashemi, Abdollah Amini, Dorsa Vatandoust, Shahin Aghamiri, Robabeh Asadi, Babak Sabet, The Combined Effects of a Methacrylate Powder Dressing (Altrazeal Powder) and Photobiomodulation Therapy on the Healing of a Severe Diabetic Foot Ulcer in a Diabetic Patient: A Case Report, 2022, 13, 2008-9783, e38, 10.34172/jlms.2022.38 | |
383. | Fernanda Cristina Toloi Rufato, Luiz Gustavo de Sousa, Priscilla Hakime Scalize, Rossano Gimenes, Isabela Hallak Regalo, Adalberto Luiz Rosa, Marcio Mateus Beloti, Fabíola Singaretti de Oliveira, Karina Fittipaldi Bombonato-Prado, Simone Cecilio Hallak Regalo, Selma Siéssere, Texturized P(VDF-TrFE)/BT membrane enhances bone neoformation in calvaria defects regardless of the association with photobiomodulation therapy in ovariectomized rats, 2022, 26, 1432-6981, 1053, 10.1007/s00784-021-04089-y | |
384. | Allan Luís Barboza Atum, José Almir Alves da Silva, Danila Marques, Renato Araújo Prates, Fernanda Marciano Consolim-Colombo, Maria Cláudia Costa Irigoyen, Maria Aparecida Dalboni, Maria Cristina Chavantes, José Antônio Silva, Photobiomodulation therapy preconditioning modifies nitric oxide pathway and oxidative stress in human-induced pluripotent stem cell-derived ventricular cardiomyocytes treated with doxorubicin, 2022, 37, 1435-604X, 1667, 10.1007/s10103-021-03416-9 | |
385. | Sajee Sattayut, Duangruthai Chotiwutthiphatthana, Todsaphon Inprakhon, Ravi Tiansongjai, An Efficacy of Photobiomodulation of 850 nm on Pain Reduction in a Human Oral Capsaicin Pain Model, 2021, 39, 2578-5478, 734, 10.1089/photob.2020.4892 | |
386. | Iuliia Golovynska, Sergii Golovynskyi, Junle Qu, Comparing the Impact of NIR , Visible and UV Light on ROS Upregulation via Photoacceptors of Mitochondrial Complexes in Normal, Immune and Cancer Cells , 2023, 99, 0031-8655, 106, 10.1111/php.13661 | |
387. | Tolga Atilla Ceranoglu, Paolo Cassano, Barbora Hoskova, Allison Green, Nina Dallenbach, Maura DiSalvo, Joseph Biederman, Gagan Joshi, Transcranial Photobiomodulation in Adults with High-Functioning Autism Spectrum Disorder: Positive Findings from a Proof-of-Concept Study, 2022, 40, 2578-5478, 4, 10.1089/photob.2020.4986 | |
388. | Guillaume Blivet, Aroa Relano-Gines, Mélanie Wachtel, Jacques Touchon, A Randomized, Double-Blind, and Sham-Controlled Trial of an Innovative Brain-Gut Photobiomodulation Therapy: Safety and Patient Compliance, 2022, 90, 13872877, 811, 10.3233/JAD-220467 | |
389. | Arteom O. Belotelov, Elena I. Cherkasova, Vladimir I. Yusupov, Nikita V. Minaev, Anastasia S. Nerush, Dmitriy V. Skamnitsky, Anna V. Maslennikova, David H. Kessel, Tayyaba Hasan, 2022, Effects of photobiomodulation on the BJ-5ta-hTERT fibroblasts exposed to ionizing radiation, 9781510647510, 29, 10.1117/12.2610227 | |
390. | Nicholas Tripodi, Fotios Sidiroglou, Vasso Apostolopoulos, Jack Feehan, Transcriptome analysis of the effects of polarized photobiomodulation on human dermal fibroblasts, 2023, 10111344, 112696, 10.1016/j.jphotobiol.2023.112696 | |
391. | Marjorie Dole, Vincent Auboiroux, Lilia Langar, John Mitrofanis, A systematic review of the effects of transcranial photobiomodulation on brain activity in humans, 2023, 0, 0334-1763, 10.1515/revneuro-2023-0003 | |
392. | Babak Arjmand, Fakher Rahim, The Probable Protective Effect of Photobiomodulation on the Immunologic Factor’s mRNA Expression Level in the Lung: An Extended COVID-19 Preclinical and Clinical Meta-analysis, 2023, 16, 2632-010X, 2632010X2211276, 10.1177/2632010X221127683 | |
393. | Laura Marinela Ailioaie, Constantin Ailioaie, Gerhard Litscher, Photobiomodulation in Alzheimer’s Disease—A Complementary Method to State-of-the-Art Pharmaceutical Formulations and Nanomedicine?, 2023, 15, 1999-4923, 916, 10.3390/pharmaceutics15030916 | |
394. | Hongya Zeng, Lan Gong, A Review of Applications and Intracellular Mechanisms of Intense Pulsed Light in Eyelid Inflammatory Diseases, 2023, 41, 2578-5478, 104, 10.1089/photob.2022.0120 | |
395. | Sara Salman, Cyprien Guermonprez, Laurent Peno-Mazzarino, Elian Lati, Audrey Rousseaud, Lieve Declercq, Saadia Kerdine-Römer, Photobiomodulation Controls Keratinocytes Inflammatory Response through Nrf2 and Reduces Langerhans Cells Activation, 2023, 12, 2076-3921, 766, 10.3390/antiox12030766 | |
396. | Doris Loh, Russel J. Reiter, Light, Water, and Melatonin: The Synergistic Regulation of Phase Separation in Dementia, 2023, 24, 1422-0067, 5835, 10.3390/ijms24065835 | |
397. | Tian Tian, Ziting Wang, Leyi Chen, Wenan Xu, Buling Wu, Photobiomodulation activates undifferentiated macrophages and promotes M1/M2 macrophage polarization via PI3K/AKT/mTOR signaling pathway, 2023, 38, 1435-604X, 10.1007/s10103-023-03753-x | |
398. | Jessica Bunch, Photobiomodulation (Therapeutic Lasers), 2023, 01955616, 10.1016/j.cvsm.2023.02.010 | |
399. | Tárik Ocon Braga Polo, João Matheus Fonseca-Santos, Gustavo Antonio Correa Momesso, William Phillip Pereira da Silva, Stefany Barbosa, Anderson Maikon de Souza Santos, Mirela Caroline Silva, Valdir Gouveia Garcia, Letícia Helena Theodoro, Leonardo P. Faverani, Single intraoperative infrared laser optimized bone repair in rat femoral osteotomies with experimentally induced osteoporosis, 2023, 38, 1435-604X, 10.1007/s10103-023-03746-w | |
400. | Ji Hyeon Ryu, Jisu Park, Bo-Young Kim, Yeonye Kim, Nam Gyun Kim, Yong-Il Shin, Photobiomodulation ameliorates inflammatory parameters in fibroblast-like synoviocytes and experimental animal models of rheumatoid arthritis, 2023, 14, 1664-3224, 10.3389/fimmu.2023.1122581 | |
401. | Mauro Célio Ribeiro Silva, Lílian Betânia Reis Amaro, Angeliny Tamiarana Lima, Alice Crespo Ferreira, Deborah de Farias Lelis, João Marcus Oliveira Andrade, André Luiz Sena Guimarães, Light-emitting diode (LED) photobiomodulation regulates thermogenesis and lipogenesis markers in adipose tissue and improves anthropometric and metabolic parameters in obese mice, 2023, 38, 1435-604X, 10.1007/s10103-023-03743-z | |
402. | Wenjun Zhang, Yun Zhao, Jianfei Dong, 2023, Impact of blue light irradiation on the viability of four types of human cells, 979-8-3503-4638-1, 259, 10.1109/SSLChinaIFWS57942.2023.10070982 | |
403. | Sung Ryeong Yoon, So-Young Chang, Min Young Lee, Jin-Chul Ahn, Effects of 660-nm LED photobiomodulation on drebrin expression pattern and astrocyte migration, 2023, 13, 2045-2322, 10.1038/s41598-023-33469-5 | |
404. | Victoria Daylor, Cortney Gensemer, Russell A. Norris, Linda Bluestein, Hope for Hypermobility: Part 2—An Integrative Approach to Treating Symptomatic Joint Hypermobility, 2023, 38, 0882-5645, 1, 10.1097/01.TPM.0000933968.28098.59 | |
405. | Joy Lodewijckx, Jolien Robijns, Marithé Claes, Maud Pierson, Melissa Lenaerts, Jeroen Mebis, The use of photobiomodulation therapy for the management of chemotherapy-induced alopecia: a randomized, controlled trial (HAIRLASER trial), 2023, 31, 0941-4355, 10.1007/s00520-023-07743-1 | |
406. | Shirin Shamloo, Erwin Defensor, Peter Ciari, Gaku Ogawa, Laura Vidano, Jennifer S. Lin, John A. Fortkort, Mehrdad Shamloo, Annelise E. Barron, The anti-inflammatory effects of photobiomodulation are mediated by cytokines: Evidence from a mouse model of inflammation, 2023, 17, 1662-453X, 10.3389/fnins.2023.1150156 | |
407. | Augustin C. Barolet, Amelia M. Villarreal, Abdulhadi Jfri, Ivan V. Litvinov, Daniel Barolet, Low-Intensity Visible and Near-Infrared Light-Induced Cell Signaling Pathways in the Skin: A Comprehensive Review, 2023, 41, 2578-5478, 147, 10.1089/photob.2022.0127 | |
408. | Euy Hyun Chung, Ji Won Son, Yun Su Eun, Na Gyeong Yang, Jae Yoon Kim, Sulhee Lee, Nam Hun Heo, Jinhui Rhee, Sung Yul Lee, Yongsung Hwang, Jung Eun Kim, Elżbieta Kłujszo, A Novel, Hand-Held, and Low-Level Light Therapy Device for the Treatment of Acne Vulgaris: A Single-Arm, Prospective Clinical Study, 2023, 2023, 1529-8019, 1, 10.1155/2023/8846620 | |
409. | Deborah R. Simkin, Post-Traumatic Stress Disorder/Developmental Trauma Disorder/Complex Post-Traumatic Stress Disorder and Complementary and Integrative Medicine/Functional Medicine, 2023, 32, 10564993, 317, 10.1016/j.chc.2022.08.011 | |
410. | A.I. Greben, P.S. Eremin, E.Yu. Kostromina, P.A. Markov, T.N. Greben, I.R. Gilmutdinova, T.V. Konchugova, Low level laser therapy: molecular mechanisms of anti-inflammatory and regenerative effects, 2023, 100, 0042-8787, 61, 10.17116/kurort202310002161 | |
411. | Latifa Mohamed Abdelgawad, Ahmed Abdullatif Abdelaziz, Mahmoud Bawdy El-Begawey, Ali Mohamed Saafan, Influence of Nanocurcumin and Photodynamic Therapy Using Nanocurcumin in Treatment of Rat Tongue Oral Squamous Cell Carcinoma Through Histological Examination and Gene Expression of BCL2 and Caspase-3, 2023, 11, 2322-3480, 730, 10.52547/rbmb.11.4.730 | |
412. | Brian Bicknell, Ann Liebert, Thomas Borody, Geoffrey Herkes, Craig McLachlan, Hosen Kiat, Neurodegenerative and Neurodevelopmental Diseases and the Gut-Brain Axis: The Potential of Therapeutic Targeting of the Microbiome, 2023, 24, 1422-0067, 9577, 10.3390/ijms24119577 | |
413. | Saina Khalaj, Babak Iranpour, Mahshid Hodjat, Arash Azizi, Mohammad Javad Kharazifard, Neda Hakimiha, Photobiomodulation effects of pulsed and continuous wave near‐infrared laser on the proliferation and migration of human gingival fibroblasts: An in vitro study, 2023, 0031-8655, 10.1111/php.13816 | |
414. | Patrick Roynard, 2023, 9781119823520, 190, 10.1002/9781119823551.ch21 | |
415. | E-Liisa Laakso, Tatjana Ewais, A Holistic Perspective on How Photobiomodulation May Influence Fatigue, Pain, and Depression in Inflammatory Bowel Disease: Beyond Molecular Mechanisms, 2023, 11, 2227-9059, 1497, 10.3390/biomedicines11051497 | |
416. | Matthew Su, Damir Nizamutdinov, Hanli Liu, Jason H. Huang, Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease, 2023, 24, 1422-0067, 9272, 10.3390/ijms24119272 | |
417. | Reham A. Shalaby, Muhammad Mohsin Qureshi, Mohd. Afzal Khan, S.M. Abdus Salam, Hyuk Sang Kwon, Kyung Hwa Lee, Euiheon Chung, Young Ro Kim, Photobiomodulation therapy restores olfactory function impaired by photothrombosis in mouse olfactory bulb, 2023, 00144886, 114462, 10.1016/j.expneurol.2023.114462 | |
418. | Giuseppina Malcangi, Assunta Patano, Irma Trilli, Fabio Piras, Anna Maria Ciocia, Alessio Danilo Inchingolo, Antonio Mancini, Denisa Hazballa, Daniela Di Venere, Francesco Inchingolo, Elisabetta de Ruvo, Gianna Dipalma, Angelo Michele Inchingolo, Therapeutic and Adverse Effects of Lasers in Dentistry: A Systematic Review, 2023, 10, 2304-6732, 650, 10.3390/photonics10060650 | |
419. | Chironjeet Chaki, Luis De Taboada, Kwong Ming Tse, Three‐dimensional irradiance and temperature distributions resulting from transdermal application of laser light to human knee—A numerical approach, 2023, 1864-063X, 10.1002/jbio.202200283 | |
420. | Tainá Caroline dos Santos Malavazzi, Kristianne Porta Santos Fernandes, Talita Christine Camilo Lopez, Maria Fernanda Setubal Destro Rodrigues, Anna Carolina Ratto Tempestini Horliana, Sandra Kalil Bussadori, Raquel Agnelli Mesquita-Ferrari, Effects of the invasive and non-invasive systemic photobiomodulation using low-level laser in experimental models: A systematic review, 2023, 38, 1435-604X, 10.1007/s10103-023-03799-x | |
421. | Denis Bouboulis, Avery Huff, Lauren Burawski, Twenty cases of perennial and seasonal allergic rhinitis treated with LumiMed® Nasal Device, 2023, 17, 1752-1947, 10.1186/s13256-023-03980-4 | |
422. | Clara Maria Cobra Branco Scontri, Flávio de Castro Magalhães, Ana Paula Moraes Damiani, Michael R. Hamblin, Antonio Roberto Zamunér, Cleber Ferraresi, Dose and time–response effect of photobiomodulation therapy on glycemic control in type 2 diabetic patients combined or not with hypoglycemic medicine: A randomized, crossover, double‐blind, sham‐controlled trial, 2023, 1864-063X, 10.1002/jbio.202300083 | |
423. | Ling Zhu, Xinpeng Gao, Ying Lv, Shuai Yu, Lu Tang, Timon Chengyi Liu, Integrated Dose–Effect Relationship of Near-Infrared Light-Emitting Diode Light on Bone Regeneration in Disuse Osteoporosis Rats, 2023, 2578-5478, 10.1089/photob.2022.0061 | |
424. | Narda G. Robinson, 2023, 9780323764698, 800, 10.1016/B978-0-323-76469-8.00145-3 | |
425. | Wuqi Song, Hanxu Zhang, Yue Pan, Qing Xia, Qiannan Liu, Hao Wu, Siqi Du, Fengmin Zhang, Hailiang Liu, LED irradiation at 630 nm alleviates collagen-induced arthritis in mice by inhibition of NF-κB-mediated MMPs production, 2023, 1474-905X, 10.1007/s43630-023-00449-7 | |
426. | Shiyang Li, Zixuan Xiao, Duyang Wang, Kainan Yang, Xianwei Cao, Guangxu Wang, Zhuxiu Guo, Jianbo Tong, Wenmin Fei, Yangyang Gong, Zhibin Zhang, Clinical efficacy of LED golden light combined with acyclovir in the treatment of herpes zoster: a single-center prospective study, 2023, 38, 1435-604X, 10.1007/s10103-023-03817-y | |
427. | Ragda Abdalla-Aslan, Yehuda Zadik, Orna Intrator, Elena Bardellini, Karis Kin Fong Cheng, Paolo Bossi, Noam Yarom, Sharon Elad, Clinical use of photobiomodulation for the prevention and treatment of oral mucositis: the real-life experience of MASCC/ISOO members, 2023, 31, 0941-4355, 10.1007/s00520-023-07919-9 | |
428. | Bruno Ferreira Botelho, Martha Carolina Torres, Ney Paredes, Aguinaldo Silva Garcez, Won Moon, Selly Sayuri Suzuki, Biomodulation of Induced Tooth Movement by Three Methods, Corticopuncture, Photobiomodulation, and Their Combination: An Animal Study, 2023, 41, 2578-5478, 328, 10.1089/photob.2022.0119 | |
429. | Jagadish Hosmani, Abdullah Alqarni, Chandrashekar Yavagal, Shahabe Saquib Abullais, Imran Khalid, Sulphi Abdul Basheer, Mohammed Ibrahim, Mohammad Y. Alshahrani, Abdulrahman Yahya Alshahrani, Photobiomodulation Therapy in Mitigating the Risk of SARS CoV-2 Infection by Enhancing Biodiversity of Oral Microbiome-A Conceptual Hypothesis Based on Scoping Review, 2023, 15, 1947-2935, 457, 10.1166/sam.2023.4493 | |
430. | Zhihao Zhang, Zhijie Zhu, Xiaoshuang Zuo, Xuankang Wang, Cheng Ju, Zhuowen Liang, Kun Li, Jiawei Zhang, Liang Luo, Yangguang Ma, Zhiwen Song, Xin Li, Penghui Li, Huilin Quan, Peipei Huang, Zhou Yao, Ning Yang, Jie Zhou, Zhenzhen Kou, Beiyu Chen, Tan Ding, Zhe Wang, Xueyu Hu, Photobiomodulation reduces neuropathic pain after spinal cord injury by downregulating CXCL10 expression, 2023, 1755-5930, 10.1111/cns.14325 | |
431. | Bruna Grazielle Quadros, Camila Marques Glen, Danielle Neres, Renata Anddresa Stachelski, Márcia Rosângela Buzanello, Gladson Ricardo Flor Bertolini, Therapeutic effects of laser on psoriasis plaques: a systematic review, 2023, 1476-4172, 1, 10.1080/14764172.2023.2241691 | |
432. | Márcia Cristina Prado Felician, Renata Belotto, João Paulo Tardivo, Mauricio S. Baptista, Waleska Kerllen Martins, Photobiomodulation: Cellular, molecular, and clinical aspects, 2023, 17, 26664690, 100197, 10.1016/j.jpap.2023.100197 | |
433. | Daisuke Uta, Naoya Ishibashi, Yuki Kawase, Shinichi Tao, Masahito Sawahata, Toshiaki Kume, Relationship between Laser Intensity at the Peripheral Nerve and Inhibitory Effect of Percutaneous Photobiomodulation on Neuronal Firing in a Rat Spinal Dorsal Horn, 2023, 12, 2077-0383, 5126, 10.3390/jcm12155126 | |
434. | Mario Migliario, Preetham Yerra, Sarah Gino, Maurizio Sabbatini, Filippo Renò, Laser Biostimulation Induces Wound Healing-Promoter β2-Defensin Expression in Human Keratinocytes via Oxidative Stress, 2023, 12, 2076-3921, 1550, 10.3390/antiox12081550 | |
435. | Farzad Salehpour, Saeed Sadigh-Eteghad, Javad Mahmoudi, Farzin Kamari, Paolo Cassano, Michael Richard Hamblin, 2023, Chapter 4, 978-3-031-36230-9, 49, 10.1007/978-3-031-36231-6_4 | |
436. | Michael R. Hamblin, Transcranial photobiomodulation for the brain: a wide range of clinical applications, 2024, 19, 1673-5374, 483, 10.4103/1673-5374.380891 | |
437. | Cassie Chin, Scott Sigman, Experience-Based Opioid-Sparing Orthopedic Surgical Protocols, 2021, 2, 2691-6541, 10.60118/001c.21694 | |
438. | Seyed Alireza Ebadi, Faraj Tabeie, Sahar Tavakoli, Shayesteh Khalili, Effects of Photobiomodulation With Two Wavelengths of 630 and 810 nm on Diabetic Neuropathy, 2023, 14, 2008-9783, e22, 10.34172/jlms.2023.22 | |
439. | Robabeh Asadi, Atarodalsadat Mostafavinia, Abdollah Amini, Houssein Ahmadi, Behnaz Ahrabi, Hamidreza Omidi, Erfan Pourhashemi, Masoumeh Hajihosseintehrani, Fatemehalsadat Rezaei, Zhaleh Mohsenifar, Sufan Chien, Mohammad Bayat, Acceleration of a delayed healing wound repair model in diabetic rats by additive impacts of photobiomodulation plus conditioned medium of adipose-derived stem cells, 2023, 2251-6581, 10.1007/s40200-023-01285-3 | |
440. | Afaf M. Botla, Jehan H. Mustafa, Amira M. Abd‐elmonem, Mohamed D. Sayed, Mai M. A. Shehata, Effect of laser acupuncture on monosymptomatic nocturnal enuresis in adolescent females: A randomized controlled trial, 2023, 1358-2267, 10.1002/pri.2048 | |
441. | Fernanda P. Santos, Carlota A. Carvalhos, Margarida Figueiredo-Dias, New Insights into Photobiomodulation of the Vaginal Microbiome—A Critical Review, 2023, 24, 1422-0067, 13507, 10.3390/ijms241713507 | |
442. | Yasmin Nikookam, Nawal Zia, Andrew Lotfallah, Jameel Muzaffar, Jennifer Davis‐Manders, Peter Kullar, Matthew Smith, Gemma Bale, Patrick Boyle, Richard Irving, Dan Jiang, Manohar Bance, The effect of photobiomodulation on hearing loss: A systematic review, 2023, 1749-4478, 10.1111/coa.14113 | |
443. | Hanieh Mohebbi, Elham Siasi, Armin Khosravipour, Mohammadali Asghari, Abdollah Amini, Atarodalsadat Mostafavinia, Mohammad Bayat, MicroRNA-26 and Related Osteogenic Target Genes Could Play Pivotal Roles in Photobiomodulation and Adipose-Derived Stem Cells-Based Healing of Critical Size Foot Defects in the Rat Model, 2023, 41, 2578-5478, 539, 10.1089/photob.2022.0128 | |
444. | Aaron Wells, Justin Rigby, Chris Castel, Dawn Castel, Pulsed Red and Blue Photobiomodulation for the Treatment of Thigh Contusions and Soft Tissue Injury: A Randomized Controlled Trial, 2023, 1056-6716, 1, 10.1123/jsr.2022-0334 | |
445. | Sonja Zarkovic Gjurin, Jason Pang, Mihael Vrčkovnik, Reem Hanna, Efficacy of 1064 nm Photobiomodulation Dosimetry Delivered with a Collimated Flat-Top Handpiece in the Management of Peripheral Facial Paralysis in Patients Unresponsive to Standard Treatment Care: A Case Series, 2023, 12, 2077-0383, 6294, 10.3390/jcm12196294 | |
446. | Radwa H. Lutfy, Sherine Abdel Salam, Haitham S. Mohammed, Marwa M Shakweer, Amina E. Essawy, Photomodulatory effects in the hypothalamus of sleep-deprived young and aged rats, 2023, 01664328, 114731, 10.1016/j.bbr.2023.114731 | |
447. | Manuela dos Santos Carvalho Schiavon, Luis Henrique Oliveira de Moraes, Thiago Francisco de Moraes, Tereza Cristina Buzinari, José Carlos Rapozo Mazulo Neto, Gerson Jhonatan Rodrigues, Chronic red laser treatment induces hypotensive effect in two-kidney one-clip model of renovascular hypertension in rat, 2023, 38, 1435-604X, 10.1007/s10103-023-03918-8 | |
448. | Blanche Aguida, Marie-Marthe Chabi, Soria Baouz, Rhys Mould, Jimmy D. Bell, Marootpong Pooam, Sebastien André, Dominique Archambault, Margaret Ahmad, Nathalie Jourdan, Near-Infrared Light Exposure Triggers ROS to Downregulate Inflammatory Cytokines Induced by SARS-CoV-2 Spike Protein in Human Cell Culture, 2023, 12, 2076-3921, 1824, 10.3390/antiox12101824 | |
449. | Camila R. Silva, Saulo T. Pereira, Daniela F.T. Silva, Lucas R. De Pretto, Anderson Z. Freitas, Carlos A. Zeituni, Maria E.C.M. Rostelato, Martha S. Ribeiro, Noninvasive Red Laser Intervention before Radiotherapy of Triple-negative Breast Cancer in a Murine Model, 2023, 200, 0033-7587, 10.1667/RADE-23-00050.1 | |
450. | Yuanhua Liu, Yongying Liang, Xiaoyuan Zhou, Jennifer E. Dent, Lucia di Nardo, Ting Jiang, Ding Qin, Youtao Lu, Dongyi He, Christine Nardini, 2023, Chapter 12, 978-3-031-34883-9, 221, 10.1007/978-3-031-34884-6_12 | |
451. | Jiawen Yong, Sabine Gröger, Julia Von Bremen, Márcia Martins Marques, Andreas Braun, Xiaoyan Chen, Sabine Ruf, Qianming Chen, Photobiomodulation therapy assisted orthodontic tooth movement: potential implications, challenges, and new perspectives, 2023, 1673-1581, 10.1631/jzus.B2200706 | |
452. | Fernanda Vieira Heimlich, José Alcides Almeida de Arruda, Nickolas Mendes Pereira, Larissa dos Santos Faria, Lucas Guimarães Abreu, Marcus Vinícius Lucas Ferreira, Fabiana Maria Kakehasi, Denise Vieira Travassos, Tarcília Aparecida Silva, Ricardo Alves Mesquita, Proposal of a prophylactic photobiomodulation protocol for chemotherapy-induced oral and oropharyngeal mucositis: a randomized clinical trial, 2023, 38, 1435-604X, 10.1007/s10103-023-03916-w | |
453. | Renlong Zhang, Junle Qu, The Mechanisms and Efficacy of Photobiomodulation Therapy for Arthritis: A Comprehensive Review, 2023, 24, 1422-0067, 14293, 10.3390/ijms241814293 | |
454. | So-Young Chang, Min Young Lee, Photobiomodulation of Neurogenesis through the Enhancement of Stem Cell and Neural Progenitor Differentiation in the Central and Peripheral Nervous Systems, 2023, 24, 1422-0067, 15427, 10.3390/ijms242015427 | |
455. | Jaimie Hoh Kam, John Mitrofanis, Glucose Improves the Efficacy of Photobiomodulation in Changing ATP and ROS Levels in Mouse Fibroblast Cell Cultures, 2023, 12, 2073-4409, 2533, 10.3390/cells12212533 | |
456. | Yuan Liu, Juan Yang, Bing Jiang, Genzi Zheng, Yao Wang, Low-energy LED red light inhibits the NF-κB pathway and promotes hPDLSCs proliferation and osteogenesis in a TNF-α environment in vitro, 2023, 38, 1435-604X, 10.1007/s10103-023-03880-5 | |
457. | Seo-Yeon Kim, Myung-Jun Song, In-Beom Kim, Tae Kwan Park, Jungmook Lyu, Photobiomodulation therapy activates YAP and triggers proliferation and dedifferentiation of Müller glia in mammalian retina, 2023, 56, 1976-670X, 502, 10.5483/BMBRep.2023-0059 | |
458. | Fábio Juner Lanferdini, Bruno Manfredini Baroni, Caetano Decian Lazzari, Raphael Luiz Sakugawa, Rodolfo André Dellagrana, Fernando Diefenthaeler, Fabrizio Caputo, Marco Aurélio Vaz, Effects of Photobiomodulation Therapy on Performance in Successive Time-to-Exhaustion Cycling Tests: A Randomized Double-Blinded Placebo-Controlled Trial, 2023, 8, 2411-5142, 144, 10.3390/jfmk8040144 | |
459. | Jie-Ren Mi Le, Jih-Huah Wu, Fu-Shan Jaw, Chuan-Tsung Su, The effect of bone remodeling with photobiomodulation in dentistry: a review study, 2023, 38, 1435-604X, 10.1007/s10103-023-03933-9 | |
460. | Breno Amaral Rocha, Polianne Alves Mendes, Giovanna Ribeiro Souto, Paulo Eduardo Alencar Souza, Soraya de Mattos Camargo Grossmann, Martinho Campolina Rebello Horta, Management of Oral Lesions in Chikungunya Virus Disease Using Photobiomodulation Therapy: A Therapy Worth Exploring, 2023, 41, 2578-5478, 595, 10.1089/photob.2023.0126 | |
461. | Sarah J. Wu, Xuanhe Zhao, Bioadhesive Technology Platforms, 2023, 0009-2665, 10.1021/acs.chemrev.3c00380 | |
462. | So-Young Chang, Eunjeong Kim, Nathaniel T. Carpena, Jae-Hun Lee, Doo Hee Kim, Min Young Lee, Ludovic Zimmerlin, Photobiomodulation Can Enhance Stem Cell Viability in Cochlea with Auditory Neuropathy but Does Not Restore Hearing, 2023, 2023, 1687-9678, 1, 10.1155/2023/6845571 | |
463. | Alaa Medhat, Medhat A. El-Zainy, Iman Fathy, Photo Biomodulation of Dental Derived Stem Cells to Ameliorate Regenerative Capacity, 2023, 10139052, 10.1016/j.sdentj.2023.11.018 | |
464. | Jakub Grzegorz Adamczyk, Support Your Recovery Needs (SYRN) – a systemic approach to improve sport performance, 2023, 15, 2080-2234, 269, 10.2478/bhk-2023-0033 | |
465. | Jing Liu, Dongyun Xia, Min Wei, Shaojing Zhou, Jian Li, Yajuan Weng, Bibliometric Analysis to Global Research Status Quo on Photobiomodulation, 2023, 2578-5478, 10.1089/photob.2023.0058 | |
466. | Goli Chamani, Mohammad Reza Zarei, Maryam Rad, Sahar Mafi, Comparison of low‐level laser therapy and standard treatment for temporomandibular disorders: An assessment of therapeutic and placebo effects, 2023, 0305-182X, 10.1111/joor.13634 | |
467. | Patricia Kasowanjete, Sathish Sundar Dhilip Kumar, Nicolette N. Houreld, A Review of Photobiomodulation on PI3K/AKT/mTOR in Wound Healing, 2023, 26664690, 100215, 10.1016/j.jpap.2023.100215 | |
468. | Tatiana V. Konchugova, Tatyana V. Apkhanova, Detelina B. Kulchitskaya, Anatoliy D. Fesyun, Maxim Yu. Yakovlev, Elena M. Styazkina, Olga M. Musaeva, Valentina А. Morunova, Olga V. Yurova, Elena A. Rozhkova, Elena S. Berezkina, Cytokine Activity Indicators Dynamics after Non-Drug Rehabilitation including Intravenous Laser Blood Irradiation in Patients with a Lower Limb Post-Thrombophlebitic Syndrome: a Randomized Study, 2023, 22, 2713-2625, 40, 10.38025/2078-1962-2023-22-3-40-48 | |
469. | Briana Paiewonsky, Margo Winter, Maria Hordinsky, MacKenzie Griffith, Ronda S. Farah, Photobiomodulation and alopecia: a crowdsourced survey study on patient preferences, 2023, 1476-4172, 1, 10.1080/14764172.2023.2284641 | |
470. | Hao Xu, Ziyi Luo, Renlong Zhang, Iuliia Golovynska, Yanxia Huang, Soham Samanta, Ting Zhou, Shaowei Li, Bingang Guo, Liwei Liu, Xiaoyu Weng, Jun He, Changrui Liao, Yiping Wang, Tymish Y. Ohulchanskyy, Junle Qu, Exploring the effect of photobiomodulation and gamma visual stimulation induced by 808 nm and visible LED in Alzheimer's disease mouse model, 2024, 250, 10111344, 112816, 10.1016/j.jphotobiol.2023.112816 | |
471. | Shaojun Liu, Dongyu Li, Tingting Yu, Jingtan Zhu, Oxana Semyachkina-Glushkovskaya, Dan Zhu, Transcranial photobiomodulation improves insulin therapy in diabetic microglial reactivity and the brain drainage system, 2023, 6, 2399-3642, 10.1038/s42003-023-05630-3 | |
472. | Ana Rita Potrich, Bruna Barcelos Só, Lauren Frenzel Schuch, Vivian Petersen Wagner, Felipe Martins Silveira, Fábio de Abreu Alves, Ana Carolina Prado-Ribeiro, Alan Roger Santos-Silva, Nathaniel Simon Treister, Manoela Domingues Martins, Marco Antonio Trevizani Martins, Impact of photobiomodulation for prevention of oral mucositis on the quality of life of patients with head and neck cancer: a systematic review, 2023, 39, 1435-604X, 10.1007/s10103-023-03940-w | |
473. | Thobekile S. Leyane, Sandy W. Jere, Nicolette N. Houreld, Effect of photobiomodulation at 830 nm on gene expression correlated with JAK/STAT signalling in wounded and diabetic wounded fibroblasts in vitro, 2023, 1864-063X, 10.1002/jbio.202300230 | |
474. | Nirosha J. Murugan, Solsa Cariba, Sawith Abeygunawardena, Nicolas Rouleau, Samantha L. Payne, Biophysical control of plasticity and patterning in regeneration and cancer, 2024, 81, 1420-682X, 10.1007/s00018-023-05054-6 | |
475. | Naoya Ishibashi, Daisuke Uta, Study of Analgesic Effects of Laser Irradiation in Rats, 2023, 0288-6200, 10.2530/jslsm.jslsm-44_0045 | |
476. | Lidong Huang, Weiyu Gong, Guibin Huang, Jingyi Li, Jilin Wu, Yanmei Dong, The additive effects of bioactive glasses and photobiomodulation on enhancing bone regeneration, 2023, 10, 2056-3426, 10.1093/rb/rbad024 | |
477. | Guo-qiang Liu, Xiao-Xuan Chen, Kun Gong, Impact of 810nm diode laser, intraoral and extraoral applications, on postoperative pain, swelling, wound healing, and patient satisfaction following mandibular third molar extraction: a comparative study, 2023, 8, 2367-2587, 10.1007/s41547-023-00206-0 | |
478. | Edna Jéssica Lima Gondim, Simony Lira Nascimento, Maria Victória Candida Gaitero, Ticiana Aparecida Alves de Mira, Andrea de Vasconcelos Gonçalves, Fernanda G Surita, Effectiveness of photobiomodulation therapy on pain intensity in postpartum women with nipple or perineal trauma: protocol for a multicentre, double-blinded, parallel-group, randomised controlled trial, 2023, 13, 2044-6055, e072042, 10.1136/bmjopen-2023-072042 | |
479. | Hyeyoon Goo, Yea-Jin Lee, Sangkeun Lee, Namgue Hong, The Anti-Inflammatory Effect of Multi-Wavelength Light-Emitting Diode Irradiation Attenuates Dry Eye Symptoms in a Scopolamine-Induced Mouse Model of Dry Eye, 2023, 24, 1422-0067, 17493, 10.3390/ijms242417493 | |
480. | Hyeyoon Goo, Min Young Lee, Yea-Jin Lee, Sangkeun Lee, Jin-Chul Ahn, Namgue Hong, Multi-Wavelength Photobiomodulation Ameliorates Sodium Iodate-Induced Age-Related Macular Degeneration in Rats, 2023, 24, 1422-0067, 17394, 10.3390/ijms242417394 | |
481. | Bardia Firouz, Lorenz Faihs, Paul Slezak, Nassim Ghaffari Tabrizi-Wizsy, Kurt Schicho, Raimund Winter, Lars Peter Kamolz, Peter Dungel, Testing the effects of photobiomodulation on angiogenesis in a newly established CAM burn wound model, 2023, 13, 2045-2322, 10.1038/s41598-023-50165-6 | |
482. | Mark Cronshaw, Valina Mylona, 2023, Chapter 7, 978-3-031-43337-5, 173, 10.1007/978-3-031-43338-2_7 | |
483. | Steven P. A. Parker, 2023, Chapter 3, 978-3-031-43337-5, 35, 10.1007/978-3-031-43338-2_3 | |
484. | Aakash Ramanishankar, Ankul Singh S, Rukaiah F Begum, Narayanan Jayasankar, Afreen Nayeem, Bhupendra G Prajapati, Shanmugasundaram Nirenjen, Unleashing light's healing power: an overview of photobiomodulation for Alzheimer's treatment, 2024, 2056-5623, 10.2144/fsoa-2023-0155 | |
485. | Candela Zorzo, Lucía Rodríguez-Fernández, Juan A. Martínez, Jorge L. Arias, Photobiomodulation increases brain metabolic activity through a combination of 810 and 660 wavelengths: a comparative study in male and female rats, 2024, 39, 1435-604X, 10.1007/s10103-023-03966-0 | |
486. | Wenqi Fu, Guo Liu, Sun‐Hun Kim, Byunggook Kim, Ok‐Su Kim, Guowu Ma, Ying Yang, Danyang Liu, Siyu Zhu, Jae‐Seok Kang, Okjoon Kim, Effects of 625 nm light‐emitting diode irradiation on preventing ER stress‐induced apoptosis via GSK‐3β phosphorylation in MC3T3‐E1, 2024, 0031-8655, 10.1111/php.13906 | |
487. | Brian Bicknell, Ann Liebert, Geoffrey Herkes, Parkinson’s Disease and Photobiomodulation: Potential for Treatment, 2024, 14, 2075-4426, 112, 10.3390/jpm14010112 | |
488. | Aline Gonçalves, Francisca Monteiro, Sofia Oliveira, Inês Costa, Susana O. Catarino, Óscar Carvalho, Jorge Padrão, Andrea Zille, Teresa Pinho, Filipe S. Silva, Optimization of a Photobiomodulation Protocol to Improve the Cell Viability, Proliferation and Protein Expression in Osteoblasts and Periodontal Ligament Fibroblasts for Accelerated Orthodontic Treatment, 2024, 12, 2227-9059, 180, 10.3390/biomedicines12010180 | |
489. | Sara Salman, Sonia Raccah, Audrey Rousseaud, Lieve Declercq, Saadia Kerdine-Römer, Pivotal roles of TRPV1 channel and Nrf2 factor in green light modulation of keratinocyte inflammatory response, 2024, 20, 26664690, 100227, 10.1016/j.jpap.2024.100227 | |
490. | Kaline de Brito Sousa, Tainá Caroline dos Santos Malavazzi, Maria Fernanda Setúbal Destro Rodrigues, Tamiris Silva, Lucas Andreo, Alessandro Melo Deana, Fabio Daumas Nunes, Sandra Kalil Bussadori, Raquel Agnelli Mesquita-Ferrari, Kristianne Porta Santos Fernandes, Effects of amber LED on inflammatory and regulatory monocytes and lymphocytes, 2024, 10111344, 112848, 10.1016/j.jphotobiol.2024.112848 | |
491. | Rasha Ahmed, Omnia Hamdy, Shaimaa Elattar, Amany Ahmed Soliman, Improving human sperm motility via red and near-infrared laser irradiation: in-vitro study, 2024, 1474-905X, 10.1007/s43630-023-00525-y | |
492. | G. A. Vostroilova, N. A. Khokhlova, D. I. Shabanov, A. A. Korchagina, D. D. Morozova, A. V. Nekrasov, Effect of interferon lambda on the generation of active oxygen species in mice under conditions of oxidative stress induced by Mitomycin C, 2024, 2782-6252, 189, 10.52419/issn2782-6252.2023.4.189 | |
493. | Yue Pan, Hanxu Zhang, Qiannan Liu, Hao Wu, Siqi Du, Wuqi Song, Fengmin Zhang, Hailiang Liu, Photobiomodulation with 630-nm LED Inhibits M1 Macrophage Polarization via STAT1 Pathway Against Sepsis-Induced Acute Lung Injury, 2024, 2578-5478, 10.1089/photob.2023.0034 | |
494. | Sana Imtiaz, Tayyab Saeed Akhter, Hamama Tul Bushra, Javeria Zahid Khan, Aasiya Niazi, Muhammad Saleem, Muhammad Umar, Effective Treatment of Solitary Rectal Ulcer Syndrome by Using Photobiomodulation Therapy: A Case Report, 2024, 2578-5478, 10.1089/photob.2023.0117 | |
495. | Deivison Santos Bonfim, Micaela Leite Fernandes, Igor Pereira Ribeiro Muniz, Ítalo Sousa Pereira, Robson Amaro Augusto da Silva, Francine Cristina da Silva, Luciano Pereira Rosa, Gabriel Pinto de Oliveira Santos, Effectiveness of 780 nm photobiomodulation as adjunct treatment for bone exposed fractures: A pilot study on radiograph, pain, and cytokines analysis, 2024, 1864-063X, 10.1002/jbio.202300348 | |
496. | Hao Lin, Dongyu Li, Jingtan Zhu, Shaojun Liu, Jingting Li, Tingting Yu, Valery V. Tuchin, Oxana Semyachkina-Glushkovskaya, Dan Zhu, Transcranial photobiomodulation for brain diseases: review of animal and human studies including mechanisms and emerging trends, 2024, 11, 2329-423X, 10.1117/1.NPh.11.1.010601 | |
497. | Mostafa Alam, Shahryar Karami, Meysam Mohammadikhah, Ashkan Badkoobeh, Mohsen Golkar, Kamyar Abbasi, Reza Sayyad Soufdoost, Lotfollah Kamali Hakim, Sahar Talebi, Reza Abdollahi Namanloo, Ahmed Hussain, Artak Heboyan, Hamid Tebyaniyan, The effect of photobiomodulation therapy in common maxillofacial injuries: Current status, 2024, 42, 0263-6484, 10.1002/cbf.3951 | |
498. | Jianshen Hu, Xiaoxi Dong, Yue Lv, Dian Hu, Duheng Fei, Huajiang Dong, Bin Liu, Hongxiao Li, Huijuan Yin, Biphasic photobiomodulation of inflammation in mouse models of common wounds, infected wounds, and diabetic wounds, 2024, 10111344, 112868, 10.1016/j.jphotobiol.2024.112868 | |
499. | Thayssa Gomes Farias, Juliana Alves Rodrigues, Márcia Soares dos Santos, Andre Luiz Mencalha, Adenilson de Souza da Fonseca, Effects of low‑power red laser and blue LED on mRNA levels from DNA repair genes in human breast cancer cells, 2024, 39, 1435-604X, 10.1007/s10103-024-04001-6 | |
500. | Nicole J. Buote, 2024, 9781119933823, 225, 10.1002/9781119933861.ch14 | |
501. | Mohammad K. Alam, Balqees Alahmari, Accelerated Orthodontic Treatment Using Photobiomodulation: A Randomized Clinical Trial, 2024, 16, 0976-4879, S543, 10.4103/jpbs.jpbs_853_23 | |
502. | L Burkow Heikkinen, M Dujovny, F Cremaschi, J Piechowski, Further advancements in the near infrared lightemitting diode: review, 2023, 8, 25749838, 173, 10.15406/ipmrj.2023.08.00355 | |
503. | Michael B. Powner, Glen Jeffery, Light stimulation of mitochondria reduces blood glucose levels, 2024, 1864-063X, 10.1002/jbio.202300521 | |
504. | Dennis Sourvanos, Timothy C. Zhu, Andreea Dimofte, Theresa M. Busch, Bradley Lander, Justin C. Burrell, Rodrigo Neiva, Joseph P. Fiorellini, A Novel Investigational Preclinical Model to Assess Fluence Rate for Dental Oral Craniofacial Tissues, 2024, 15721000, 104015, 10.1016/j.pdpdt.2024.104015 | |
505. | Charles McGlynn, Doni Dermawan, Guia Marie Canonizado, Neuron Foundry Mach 8: A Concept of Multi-modal Neuro-supportive Energy Therapy Device for Treating Neurodegenerative Disorders, 2024, 2096-5958, 10.26599/BSA.2023.9050026 | |
506. | Laiz Moreira de Paula, Alessah Carolyna de Andrade Fernandes, Beatriz Castro Evangelista, Francine do Couto Lima-Moreira, Geovana Andrade, João Victor de Andrade Fernandes, Fabrício Luscino Alves de Castro, Virgílio Moreira Roriz, Clinical and thermographic evaluation after lower third molar extractions and the application of different photobiomodulation protocols: double-blind randomised clinical trial, 2024, 28, 1436-3771, 10.1007/s00784-024-05594-6 | |
507. | Marcele Florêncio das Neves, Ana Paula Pinto, Letícia Tiemi Maegima, Fernanda Pupio Silva Lima, Rodrigo Álvaro Brandão Lopes-Martins, Emilia Angela Lo Schiavo Arisawa, Mário Oliveira Lima, Effects of photobiomodulation on pain, lactate and muscle performance (ROM, torque, and EMG parameters) of paretic upper limb in patients with post-stroke spastic hemiparesis—a randomized controlled clinical trial, 2024, 39, 1435-604X, 10.1007/s10103-024-04035-w | |
508. | Sara Muñoz Declara, Aldo D’Alessandro, Agnese Gori, Benedetta Cerasuolo, Sonia Renzi, Michele Berlanda, Eric Zini, Monica Monici, Duccio Cavalieri, Giordana Zanna, Evaluation of the Impact of Near-Infrared Multiwavelength Locked System Laser Therapy on Skin Microbiome in Atopic Dogs, 2024, 14, 2076-2615, 906, 10.3390/ani14060906 | |
509. | David Richer Araujo Coelho, Joshua D. Salvi, Willians Fernando Vieira, Paolo Cassano, Inflammation in obsessive–compulsive disorder: A literature review and hypothesis‐based potential of transcranial photobiomodulation, 2024, 102, 0360-4012, 10.1002/jnr.25317 | |
510. | Anna Sancho-Balsells, Sara Borràs-Pernas, Francesca Flotta, Wanqi Chen, Daniel del Toro, Manuel J. Rodríguez, Jordi Alberch, Guillaume Blivet, Jacques Touchon, Xavier Xifró, Albert Giralt, Brain–gut photobiomodulation restores cognitive alterations in chronically stressed mice through the regulation of Sirt1 and neuroinflammation, 2024, 01650327, 10.1016/j.jad.2024.03.075 | |
511. | Ping Lu, Jinfeng Peng, Jie Liu, Lili Chen, The role of photobiomodulation in accelerating bone repair, 2024, 188, 00796107, 55, 10.1016/j.pbiomolbio.2024.03.002 | |
512. | Quentin Perrier, Cécile Moro, Sandrine Lablanche, Diabetes in spotlight: current knowledge and perspectives of photobiomodulation utilization, 2024, 15, 1664-2392, 10.3389/fendo.2024.1303638 | |
513. | Julia A. Scott, Akshat Kalra, Devanshi Pratiher, Marvin Berman, Sally Wood, James D. Carroll, Ann Liebert, Jeri-Anne Lyons, 2024, Design and validation of EEG biomarkers for transcranial photobiomodulation, 9781510669116, 25, 10.1117/12.3021832 | |
514. | Ting-Jiun Chen, Bartosz Kula, Maria Kukley, 2024, Chapter 15, 978-1-0716-3741-8, 313, 10.1007/978-1-0716-3742-5_15 | |
515. | Febriyanti Puspitasari, Akhmad Taufiq Mukti, A Shofy Mubarak, Pengaruh Penembakan Laser Inframerah pada Titik Reproduksi Terhadap Tingkat Kematangan Gonad, Fertilitas, Daya Tetas Telur dan Sitasan Larva Ikan Patin Siam Betina, 2023, 14, 2716-2702, 118, 10.30736/grouper.v14i2.211 | |
516. | Mandeep Kumar Jangra, Mehroz Ahmed Mirza, Manu Goyal, Class IV Laser Therapy as an Adjunct to Primary Percutaneous Coronary Intervention in Individuals with Acute Coronary Syndrome: A Feasibility Pilot Study, 2024, 8, 2588-9834, 87, 10.4103/bbrj.bbrj_286_23 | |
517. | 2024, 9781119879541, 363, 10.1002/9781119879558.ch6 | |
518. | 2024, 9781119879541, 503, 10.1002/9781119879558.ch9 | |
519. | 2024, 9781119879541, 773, 10.1002/9781119879558.app1 | |
520. | Daisuke Uta, Naoya Ishibashi, Shinichi Tao, Masahito Sawahata, Toshiaki Kume, Photobiomodulation inhibits neuronal firing in the superficial but not deep layer of a rat spinal dorsal horn, 2024, 710, 0006291X, 149873, 10.1016/j.bbrc.2024.149873 | |
521. | Steven Parker, Mark Cronshaw, Eugenia Anagnostaki, Valina Mylona, Edward Lynch, Martin Grootveld, Effect of Photobiomodulation Therapy Dosage on Orthodontic Movement, Temporomandibular Dysfunction and Third Molar Surgery Outcomes: A Five-Year Systematic Review, 2024, 14, 2076-3417, 3049, 10.3390/app14073049 | |
522. | Maria Luisa Hernández-Bule, Jorge Naharro-Rodríguez, Stefano Bacci, Montserrat Fernández-Guarino, Unlocking the Power of Light on the Skin: A Comprehensive Review on Photobiomodulation, 2024, 25, 1422-0067, 4483, 10.3390/ijms25084483 | |
523. | Wiktoria Julia Krzesłowska, Anna Woźniacka, The Frontal Fibrosing Alopecia Treatment Dilemma, 2024, 13, 2077-0383, 2137, 10.3390/jcm13072137 | |
524. | Jiangong Zhang, Jiahuan Liu, Yang Huang, Linlin Yan, Shufeng Xu, Guozheng Zhang, Lei Pei, Huachen Yu, Xisong Zhu, Xiaowei Han, Current role of magnetic resonance imaging on assessing and monitoring the efficacy of phototherapy, 2024, 0730725X, 10.1016/j.mri.2024.04.012 | |
525. | Elaine Aparecida Rocha Domingues, Lana Luiza da Cruz Silva, Marcelly Silva Dourado, Cintia Teles de Argôlo, Combination of phototherapy for treatment of labial necrotizing lesion, 2024, 23, 1677-3225, e242214, 10.20396/bjos.v23i00.8672214 | |
526. | Milad Iravani, Abbas Ebrahimi Kalan, Maryam Moghaddam Salimi, Ali Jahan, The Effect of Transcranial Photobiomodulation for Motor Performance Improvement in Patients with Brain Disorders, 2024, 19, 15743624, 10.2174/0115743624250965231116060824 | |
527. | Andrew R. Stevens, Mohammed Hadis, Alice Phillips, Abhinav Thareja, Michael Milward, Antonio Belli, William Palin, David J. Davies, Zubair Ahmed, Implantable and transcutaneous photobiomodulation promote neuroregeneration and recovery of lost function after spinal cord injury, 2024, 2380-6761, 10.1002/btm2.10674 | |
528. | Maria da Glória B de Melo, Luciane H Azevedo, Luiz Fernando N Ruiz, Maristela M Lobo, Patrícia M de Freitas, Photobiomodulation Therapy in the Management of Late Complications After Facial Filling, 2024, 2168-8184, 10.7759/cureus.59513 | |
529. | Yax Cuellar, Sergio Andrés Velásquez, Angela Domínguez, Root Regeneration with Photobiomodulation of an Upper Lateral Incisor Associated with Root Resorption Due to an Impacted Maxillary Canine: A Case Report, 2024, 2578-5478, 10.1089/photob.2023.0150 | |
530. | Arun Sachdev, Improvement in Central Serous Chorioretinopathy Following Multiwavelength Photobiomodulation Treatment – Case Report, 2024, 2193-8245, 10.1007/s40123-024-00963-6 | |
531. | Hui Ding, Xiao-chun Chen, Lin Wan, Ying-ying Zhang, Xiao-hong Rui, Tian He, Jun Liu, Zhong-bo Shang, Klebsiella pneumoniae alters zebrafish circadian rhythm via inflammatory pathways and is dependent on light cues, 2024, 10, 24058440, e30829, 10.1016/j.heliyon.2024.e30829 | |
532. | S. D. Astuti, R. Nashichah, P. Widiyanti, E. M. Setiawatie, M. S. Amir, A. Apsari, . Widyastuti, E. Hermanto, Y. Susilo, A. K. Yaqubi, D.Z. I. Nurdin, N. Anuar, Effectiveness of 650 nm red laser photobiomodulation therapy to accelerate wound healing post tooth extraction, 2024, 13, 2413-9432, 4, 10.24931/2413-9432-2024-13-1-4-15 | |
533. | H. H. Soltan, A. Afifi, A. Mahmoud, M. Refaat, O. F. Al Balah, Effects of silver nanoparticle and low-level laser on the immune response and healing of albino mice skin wounds, 2024, 13, 2413-9432, 16, 10.24931/2413-9432-2023-13-1-16-27 | |
534. | Naoya Ishibashi, Takuya Nanjo, Shinichi Tao, Photobiomodulation improves acute restraint stress-induced visceral hyperalgesia in rats, 2024, 39, 1435-604X, 10.1007/s10103-024-04091-2 | |
535. | Richard M. Morgan, Tyler D. Wheeler, Mark A. Poolman, Erin N. J. Haugen, Steven D. LeMire, John S. Fitzgerald, Effects of Photobiomodulation on Pain and Return to Play of Injured Athletes: A Systematic Review and Meta-analysis, 2024, 38, 1064-8011, e310, 10.1519/JSC.0000000000004752 | |
536. | Larissa Alexsandra da Silva Neto Trajano, Priscyanne Barreto Siqueira, Mariana Moreno de Sousa Rodrigues, Bruno Ricardo Barreto Pires, Adenilson de Souza da Fonseca, Andre Luiz Mencalha, Does photobiomodulation alter mitochondrial dynamics?, 2024, 0031-8655, 10.1111/php.13963 | |
537. | Beng Jiong Ang, Nursakinah Suardi, Muhammad Asyraf Abduraman, Exploring differentiation-dependent responses to 532 nm green laser photobiomodulation in SHSY5Y neuroblastoma cells, 2024, 39, 1435-604X, 10.1007/s10103-024-04102-2 | |
538. | A. A. Lau, K. Jin, H. Beard, T. Windram, K. Xie, J. A. O’Brien, D. Neumann, B. M. King, M. F. Snel, P. J. Trim, J. Mitrofanis, K. M. Hemsley, P. J. Austin, Photobiomodulation in the infrared spectrum reverses the expansion of circulating natural killer cells and brain microglial activation in Sanfilippo mice, 2024, 0022-3042, 10.1111/jnc.16145 | |
539. | Tahsin Nairuz, Jong-Ha Lee, Photobiomodulation Therapy on Brain: Pioneering an Innovative Approach to Revolutionize Cognitive Dynamics, 2024, 13, 2073-4409, 966, 10.3390/cells13110966 | |
540. | Andrew E.-Y. Chuang, Pei-Wei Weng, Chia-Hung Liu, Pei-Ru Jheng, Lekshmi Rethi, Hieu Trung Nguyen, Hsien-Tsung Lu, Photo-responsive platelet vesicles-iron oxide nanoparticles-bioglass composite for orthopedic bioengineering, 2024, 02638223, 118263, 10.1016/j.compstruct.2024.118263 | |
541. | Dongqi Liu, Xiao Luo, Yin Jing, Tingting Wu, Fangchun Chen, The Application of PBM Therapy in Patients Subjected to Oral GVHD: A Review, 2024, 2578-5478, 10.1089/pho.2023.0153 | |
542. | Piyali Bhattacharjee, Andrea Szabo, Peter Dungel, Christina Streli, Andreas Walter, 2024, Chapter 12-1, 978-3-030-85569-7, 1, 10.1007/978-3-030-85569-7_12-1 | |
543. | Paula Caroline Garcia, Danilo Peron Meireles, João Alberto Fioravante Tassinary, Christiane Pavani, Combining red photobiomodulation therapy with polydioxanone threads for wrinkle reduction in the glabella region: A randomized, controlled, double‐blind clinical trial, 2024, 1864-063X, 10.1002/jbio.202400139 | |
544. | Kyung Jin Seo, Jung Hwan Yoon, Bom Yee Chung, Hae Kyung Lee, Won Sang Park, Hiun Suk Chae, Effects of photobiomodulation on colon cancer cell line HT29 according to mitochondria, 2024, 10111344, 112966, 10.1016/j.jphotobiol.2024.112966 | |
545. | I. Kalampouka, R. R. Mould, S. W. Botchway, A. M. Mackenzie, A. V. Nunn, E. L. Thomas, J. D. Bell, Selective induction of senescence in cancer cells through near‐infrared light treatment via mitochondrial modulation, 2024, 1864-063X, 10.1002/jbio.202400046 | |
546. | Rosani Teresa de Siqueira, Luiz Claudio de Freitas, Thomas Stravinskas Durigon, Lara Jansiski Motta, Sandra Kalil Bussadori, Anna Carolina Ratto Tempestini Horliana, Raquel Agnelli Mesquita-Ferrari, Aldo Brugnera Júnior, Kristianne Porta Santos Fernandes, Cinthya Cosme Gutierrez Duran, Case Report: Photobiomodulation improves shoulder functionality after acute traumatic anterior dislocation associated with Hill-Sachs bone lesion’, 2024, 13, 2046-1402, 772, 10.12688/f1000research.152487.1 | |
547. | 2024, 9781394237951, 257, 10.1002/9781394237999.ch13 | |
548. | Gisele Dias da Silva, Fabiana Lessa Silva, Anaiá da Paixão Sevá, Danilo Machado Deorce, Nerildo de Jesus da Costa Junior, Fernanda Amaral Silva, Fernando Alzamora Filho, Effect of combined red and infrared wavelengths on inflammation, hemorrhage, and muscle damage caused by Bothrops leucurus snake venom, 2024, 39, 1435-604X, 10.1007/s10103-024-04116-w | |
549. | Daisuke Uta, Naoya Ishibashi, Study of the Mechanisms of Analgesic by Photobiomodulation—Establishment of Electrophysiological Analysis by Using Animals—, 2024, 0288-6200, 10.2530/jslsm.jslsm-45_0024 | |
550. | Yaieo Koang, A meta-analysis on the use of photobiomodulation to regulate gingival wound healing in addition to periodontal therapies, 2024, 2, 3005-480X, 107, 10.61466/ijcmr2040002 | |
551. | Jeanne Karlette Merlo, Adriano Valmozino da Silva, Juliano Casonatto, Alex Silva Ribeiro, Eros de Oliveira Junior, Ana Paula do Nascimento, Raphael Gonçalves de Oliveira, Cosme Franklim Buzzachera, Rubens Alexandre da Silva, Andreo Fernando Aguiar, Effects of a Mat Pilates Exercise Program Associated with Photobiomodulation Therapy in Patients with Chronic Nonspecific Low Back Pain: A Randomized, Double-Blind, Sham-Controlled Trial, 2024, 12, 2227-9032, 1416, 10.3390/healthcare12141416 | |
552. | Rya Cornelia Holzman, Pro-inflammatory cytokine activity: The root cause of catamenial seizures, 2024, 03069877, 111441, 10.1016/j.mehy.2024.111441 | |
553. | Reem Hanna, Ioana Cristina Miron, Snehal Dalvi, Praveen Arany, René Jean Bensadoun, Stefano Benedicenti, A Systematic Review of Laser Photobiomodulation Dosimetry and Treatment Protocols in the Management of Medications-Related Osteonecrosis of the Jaws: A Rationalised Consensus for Future Randomised Controlled Clinical Trials, 2024, 17, 1424-8247, 1011, 10.3390/ph17081011 | |
554. | Parizotto Nivaldo Antonio, Ferraresi Cleber , Enhancing Physiotherapy Outcomes with Photobiomodulation: A Comprehensive Review, 2024, 8, 25736264, 031, 10.29328/journal.jnpr.1001061 | |
555. | Faraj Tabeie, Sara Tabakhha, Soheila Sadeghi, Alireza Ebadi, Effect of Visible and Infrared Photobiomodulation on Diabetic Foot Ulcers, 2024, 15, 2008-9783, e12, 10.34172/jlms.2024.12 | |
556. | Patrícia Fernandes do Prado, Edna de Freitas Gomes Ruas, Aurelina Gomes e Martins, Simone Guimarães Teixeira Souto, Ely Carlos Pereira de Jesus, Hugo Emanuel Santos Pimenta, Fernandez Fonseca Almeida, Carla Silvana de Oliveira e Silva, Effect of the use of low power laser in the treatment of chronic wounds: an integrative review, 2024, 16, 1989-4155, e4943, 10.55905/cuadv16n7-124 | |
557. | Ali Moradi, Marefat Ghaffari Novin, Mohammad Bayat, A Comprehensive Systematic Review of the Effects of Photobiomodulation Therapy in Different Light Wavelength Ranges (Blue, Green, Red, and Near-Infrared) on Sperm Cell Characteristics in Vitro and in Vivo, 2024, 1933-7191, 10.1007/s43032-024-01657-x | |
558. | Shijing Wang, Letian Chen, Zheng Ma, Liting Zhao, Yueying Lu, Yuming Fu, Hong Liu, Gut microbiota mediates the anti‐inflammatory effects of supplemental infrared irradiation in mice, 2024, 0031-8655, 10.1111/php.14008 | |
559. | Siriluk Thammasart, Poommaree Namchaiw, Kwanchanok Pasuwat, Khaow Tonsomboon, Anak Khantachawana, Neuroprotective Potential of Photobiomodulation Therapy: Mitigating Amyloid-Beta Accumulation and Modulating Acetylcholine Levels in an In Vitro Model of Alzheimer’s Disease, 2024, 2578-5478, 10.1089/pho.2024.0042 | |
560. | Bruna Silva Gomes, Amanda Bezerra Gonçalves, Sabrina Zanchetta Lanza, Marcelo Augusto Marretto Esquisatto, Fernando Russo Costa do Bomfim, Gaspar de Jesus Lopes Filho, Effects of Photobiomodulation Associated with Platelet-Rich Plasma in Acute Rheumatoid Arthritis Induced in Female Wistar Rats’ Knee, 2024, 2578-5478, 10.1089/pho.2024.0060 | |
561. | Jianhua Huang, Qing Fan, Lei Shi, Jie Shen, Hongwei Wang, A novel chlorin derivative Shengtaibufen (STBF) mediated photodynamic therapy combined with iodophor for the treatment of chronic superficial leg wounds infected with methicillin-resistant Staphylococcus aureus: A retrospective clinical study, 2024, 15721000, 104300, 10.1016/j.pdpdt.2024.104300 | |
562. | Carla S. Sousa, Andreia Monteiro, António J. Salgado, Nuno A. Silva, Combinatorial therapies for spinal cord injury repair, 2025, 20, 1673-5374, 1293, 10.4103/NRR.NRR-D-24-00061 | |
563. | Luísa Belluco Guerrini, Nicole Rosa de Freitas, Lucas José de Azevedo-Silva, Caroline Chapernate Vieira dos Santos, Maria Aparecida de Andrade Moreira Machado, Thais Marchini de Oliveira, Ana Lúcia Pompéia Fraga de Almeida, Clinical Study of Photobiomodulation Effect at Postoperative Clefted Alveolar Ridge Bone Grafting, 2024, 1049-2275, 10.1097/SCS.0000000000010524 | |
564. | Thawatchai Thoradit, Marthe Chabi, Blanche Aguida, Soria Baouz, Verene Stierle, Marootpong Pooam, Stephane Tousaints, Casimir D. Akpovi, Margaret Ahmad, Hypersensitivity to man-made electromagnetic fields (EHS) correlates with immune responsivity to oxidative stress: a case report, 2024, 17, 1942-0889, 10.1080/19420889.2024.2384874 | |
565. | Antoinette Antwi, Alexander W. Schill, Rachel Redfern, Eric R. Ritchey, Effect of low‐level light therapy in individuals with dry eye disease, 2024, 0275-5408, 10.1111/opo.13371 | |
566. | Rongan Ye, Yu He, Wei Ni, Yiqiu Zhang, Ying Zhu, Muqing Cao, Ruida He, Min Yao, LLLT accelerates experimental wound healing under microgravity conditions via PI3K/AKT-CCR2 signal axis, 2024, 12, 2296-4185, 10.3389/fbioe.2024.1387474 | |
567. | Marucia Chacur, Igor R Correia Rocha, Michael E. Harland, Suzanne M. Green-Fulgham, Sonia Regina Yokomizo de Almeida, Adriano Polican Ciena, Linda R. Watkins, Prevention and reversal of neuropathic pain by near-infrared photobiomodulation therapy in male and female rats, 2024, 286, 00319384, 114680, 10.1016/j.physbeh.2024.114680 | |
568. | Theodore A. Henderson, Can infrared light really be doing what we claim it is doing? Infrared light penetration principles, practices, and limitations, 2024, 15, 1664-2295, 10.3389/fneur.2024.1398894 | |
569. | Saghil Sali, Rifai Chai, Balasankar Ganesan, 2025, 9780443222702, 99, 10.1016/B978-0-443-22270-2.00007-1 | |
570. | Augustin C. Barolet, Lucie Germain, Daniel Barolet, In vivo measurement of nitric oxide release from intact human skin post photobiomodulation using visible and near-infrared light: A chemiluminescence detection study, 2024, 24, 26664690, 100250, 10.1016/j.jpap.2024.100250 | |
571. | José Carlos Tatmatsu-Rocha, Luan Santos Mendes-Costa, Inflammatory markers, oxidative stress, and mitochondrial dynamics: Repercussions on coronary artery disease in diabetes, 2024, 15, 1948-9358, 1853, 10.4239/wjd.v15.i9.1853 | |
572. | Fazal S Mujawar, Sameer A Zope, Girish Suragimath, Siddhartha Varma, Apurva V Kale, Salivary Macrophage Inflammatory Protein-1α Levels in Periodontitis Subjects Receiving Non-surgical Periodontal Therapy With and Without Photo-Biomodulation: A Prospective Interventional Controlled Trial, 2024, 2168-8184, 10.7759/cureus.68980 | |
573. | Vander Oliveira de Andrade Filho, Marina Oliveira Coura Amarante, Francisco Gonzalez-Lima, Sérgio Gomes da Silva, Fabrízio dos Santos Cardoso, Systematic review of photobiomodulation for multiple sclerosis, 2024, 15, 1664-2295, 10.3389/fneur.2024.1465621 | |
574. | Ali A. Khalifa, Mohamed M.A. Ibrahim, Mahmoud S.A. Abdelrahman, Treatment of dry eye disease due to meibomian gland dysfunction using intense pulsed light and low-level light therapy, 2021, 22, 1110-9173, 91, 10.4103/djo.djo_68_20 | |
575. | Daniel Ziental, Beata Czarczynska-Goslinska, Marcin Wysocki, Marcin Ptaszek, Łukasz Sobotta, Advances and perspectives in use of semisolid formulations for photodynamic methods, 2024, 09396411, 114485, 10.1016/j.ejpb.2024.114485 | |
576. | Peng-Fei Cheng, Meng-Meng Ge, Da-Wei Ye, Jian-Ping Chen, Jin-Xi Wang, Targeting the Main Sources of Reactive Oxygen Species Production: Possible Therapeutic Implications in Chronic Pain, 2024, 22, 1570159X, 1960, 10.2174/1570159X22999231024140544 | |
577. | Jéssica de Oliveira Rossi, Emilie Maria Cabral Araujo, Maria Eduarda Côrtes Camargo, Rui Seabra Ferreira Junior, Benedito Barraviera, Maria Angélica Miglino, Dayane Maria Braz Nogueira, Carlos Henrique Bertoni Reis, Guilherme Eugênio Gil, Thaís Rissato Vinholo, Thiago Pereira Soares, Rogerio Leone Buchaim, Daniela Vieira Buchaim, Effectiveness of the Association of Fibrin Scaffolds, Nanohydroxyapatite, and Photobiomodulation with Simultaneous Low-Level Red and Infrared Lasers in Bone Repair, 2024, 17, 1996-1944, 4351, 10.3390/ma17174351 | |
578. | Julia Lawrence, Karin Sorra, Photobiomodulation as Medicine: Low-Level Laser Therapy (LLLT) for Acute Tissue Injury or Sport Performance Recovery, 2024, 9, 2411-5142, 181, 10.3390/jfmk9040181 | |
579. | Gabriela Silva, Saulo Soares da Silva, Dimitrius Santiago Passos Simões Fróes Guimarães, Marcos Vinicius da Cruz, Leonardo Reis Silveira, Etel Rocha-Vieira, Fabiano Trigueiro Amorim, Flavio de Castro Magalhaes, The dose-effect response of combined red and infrared photobiomodulation on insulin resistance in skeletal muscle cells, 2024, 40, 24055808, 101831, 10.1016/j.bbrep.2024.101831 | |
580. | Manuel Alejandro Herrera, Arthur Pereira Ribas, Paulo Eduardo da Costa, Mauricio S. Baptista, Red-light photons on skin cells and the mechanism of photobiomodulation, 2024, 5, 2673-6853, 10.3389/fphot.2024.1460722 | |
581. | Toshihiro Kushibiki, 2024, Chapter 10, 978-981-97-4255-4, 189, 10.1007/978-981-97-4256-1_10 | |
582. | Daniella Silva Oggiam, Juliana Vallim Jorgetto, Guilherme Luiz Chinini, Mônica Antar Gamba, Denise Miyuki Kusahara, Effects of Monochromatic Infrared Light on Painful Diabetic Polyneuropathy: Randomized Controlled Trial, 2024, 15249042, 10.1016/j.pmn.2024.08.012 | |
583. | Kamila Pasternak-Mnich, Jolanta Kujawa, Justyna Agier, Elżbieta Kozłowska, Impact of photobiomodulation therapy on pro-inflammation functionality of human peripheral blood mononuclear cells – a preliminary study, 2024, 14, 2045-2322, 10.1038/s41598-024-74533-y | |
584. | Esmaeil Mohammadzadeh, Aref Hosseinian Amiri, Reza Fekrazad, Rainer A. Leitgeb, Winfried Mayr, Kamran Ezzati, The Effect of Photobiomodulation on Bone Mineral Density, Serum Vitamin D, and Bone Formation Markers in Individuals with Complete Spinal Cord Injuries with Osteoporosis, 2024, 2578-5478, 10.1089/photob.2023.0195 | |
585. | So-Young Chang, Min Young Lee, Photobiomodulation as a Potential Adjuvant Therapy to Improve Cochlear Implant Efficiency, 2024, 2578-5478, 10.1089/photob.2024.0097 | |
586. | Akila Weerasekera, David Richer Araujo Coelho, Eva-Maria Ratai, Katherine Anne Collins, Aura Maria Hurtado Puerto, Luis De Taboada, Maia Beth Gersten, Julie A Clancy, Matthew J Hoptman, Molly Kennedy Irvin, Allison Mary Sparpana, Elizabeth F Sullivan, Xiaotong Song, Arwa Adib, Paolo Cassano, Dan Vlad Iosifescu, Dose-dependent effects of transcranial photobiomodulation on brain temperature in patients with major depressive disorder: a spectroscopy study, 2024, 39, 1435-604X, 10.1007/s10103-024-04198-6 | |
587. | Zahra AL-Timimi, Examining the combined benefits of photobiomodulation and apigenin for the treatment of asthenozoospermia: An innovative therapeutic strategy, 2024, 1474-905X, 10.1007/s43630-024-00643-1 | |
588. | Thayssa Gomes Farias, Márcia Soares dos Santos, Andre Luiz Mencalha, Adenilson de Souza da Fonseca, Low-power red laser and blue LED modulate telomere maintenance and length in human breast cancer cells, 2024, 39, 1435-604X, 10.1007/s10103-024-04194-w | |
589. | Bibhus Luitel, Tanush Duggisani, Anuj Luitel, John LaRocco, Reviewing the efficiency of photobiomodulation therapy in oncological treatment, 2024, 14, 2234-943X, 10.3389/fonc.2024.1447653 | |
590. | Jingfei Fu, Rui Zhao, Yiyang Jiang, Yingyi Chen, Juan Du, Yi Liu, Junji Xu, Photobiomodulation suppresses allergic contact dermatitis by inhibiting T‐cell activation, 2024, 0105-1873, 10.1111/cod.14713 | |
591. | Ann Liebert, Brian Bicknell, E-Liisa Laakso, Sharon Tilley, Gillian Heller, Hosen Kiat, Geoffrey Herkes, Improvements in clinical signs and symptoms of Parkinson’s disease using photobiomodulation: a five-year follow-up, 2024, 24, 1471-2377, 10.1186/s12883-024-03857-z | |
592. | Jorden Xavier, Daniel Grande, Seth Sherman, Kenneth Zaslav, James Paci, Investigating the Efficacy of Bioactive Sleeves with Embedded Nano-Semiconductors in Alleviating Tendinopathy: An In Vivo Pilot Study, 2024, 5, 2691-6541, 10.60118/001c.121294 | |
593. | Jumana Alsarhan, Rita El Feghali, Thaer Alkhudari, Stefano Benedicenti, Non-Pharmacological Therapies for Management of Temporomandibular Myofascial Pain Syndrome: Laser Photobiomodulation or Dry Needling? Meta-Analyses of Human Clinical Trials, 2024, 11, 2304-6732, 965, 10.3390/photonics11100965 | |
594. | Jalal Maghfour, David M. Ozog, Jessica Mineroff, Jared Jagdeo, Indermeet Kohli, Henry W. Lim, Photobiomodulation CME part I: Overview and mechanism of action, 2024, 91, 01909622, 793, 10.1016/j.jaad.2023.10.073 | |
595. | Juliana Sales Rodrigues Costa, Gabriela Silva, Isabela Carvalho Guimarães, Luis Filipe Rocha Silva, Saulo Soares da Silva, João Paulo de Paula Almeida, Cândido Celso Coimbra, Nivaldo Antonio Parizotto, Fernando Gripp, Marco Fabrício Dias‐Peixoto, Elizabethe Adriana Esteves, Fabiano Trigueiro Amorim, Cleber Ferraresi, Flavio de Castro Magalhaes, Photobiomodulation Enhances the Effect of Strength Training on Insulin Resistance Regardless of Exercise Volume in Mice Fed a High‐Fat Diet, 2024, 1864-063X, 10.1002/jbio.202400274 | |
596. | Silvana Torres Perez, Lucas Andreo, José Antonio Silva Junior, Sandra Kalil Bussadori, Anna Carolina Ratto Tempestini Horliana, Raquel Agnelli Mesquita‐Ferrari, Kristianne Porta Santos Fernandes, Effect of Photobiomodulation on an Experimental Model of Lower Limb Ischemia, 2024, 1864-063X, 10.1002/jbio.202400305 | |
597. | Chrysi Pouliou, Christina Piperi, Advances of Oxidative Stress Impact in Periodontitis: Biomarkers and Effective Targeting Options, 2024, 31, 09298673, 6187, 10.2174/0109298673297545240507091410 | |
598. | Maria Zaharieva Mutafchieva, Milena Nenkova Draganova, Blagovesta Konstantinova Yaneva, Plamen Ivanov Zagorchev, Georgi Tomchev Tomov, Clinical Improvement and P63-Deficiency Correction in OLP Patients After Photobiomodulation, 2024, 12, 2304-6767, 338, 10.3390/dj12110338 | |
599. | Fazele Atarbashi‐Moghadam, Amirhosein Mahmoudian, Niloofar Taghipour, Neda Hakimiha, Ali Azadi, Hanieh Nokhbatolfoghahaei, Enhancement of the angiogenic differentiation in the periodontal ligament stem cells using fibroblast growth factor 2 and photobiomodulation: An in vitro investigation, 2024, 0031-8655, 10.1111/php.14032 | |
600. | Caroline Covatti, Daniela Sayuri Mizobuti, Guilherme Luiz da Rocha, Heloina Nathalliê Mariano da Silva, Elaine Minatel, Photobiomodulation Therapy Effects at Different Stages of the Dystrophic Phenotype: A Histomorphometric Study, 2024, 01614754, 10.1016/j.jmpt.2024.09.008 | |
601. | Quentin Perrier, Cécile Cottet-Rousselle, Fréderic Lamarche, Emily Tubbs, Cindy Tellier, Jade Veyrat, Guillaume Vial, Pierre Bleuet, Aude Durand, Amandine Pitaval, Marie-Line Cosnier, Cécile Moro, Sandrine Lablanche, Long-term safety of photobiomodulation exposure to beta cell line and rat islets in vitro and in vivo, 2024, 14, 2045-2322, 10.1038/s41598-024-77660-8 | |
602. | Larissa Trarbach Figueiredo Braga, Isadora Martins Ribeiro, Maria Eduarda de Souza Barroso, Edgar Hell Kampke, Lorena Nascimento Santos Neves, Sara Cecília Andrade, Guilherme Heleodoro Barbosa, Marcella Leite Porto, Silvana Santos Meyrelles, Modulatory Effects of Photobiomodulation on Oxidative and Inflammatory Responses in a Murine Model of Periodontitis, 2024, 13, 2076-3921, 1450, 10.3390/antiox13121450 | |
603. | Maíra da Silva Almeida Rocha, Lucas Renan Sena de Oliveira, Diêgo Mendes Xavier, Natielle Cecília dos Santos Ottone, Pedro Paulo Ribeiro Ferreira, Kaio Augusto Oliveira Paulino, Juliana Sales Rodrigues Costa, Graciene Fernandes Araújo Campos Fonseca, Gabriela Silva, Patrick Almeida Silva, Saulo Soares da Silva, João Paulo de Paula Almeida, Cândido Celso Coimbra, Elizabethe Adriana Esteves, Cleber Ferraresi, Flavio de Castro Magalhaes, The effects of different doses of single‐ and dual‐wavelength whole‐body photobiomodulation on metabolic parameters in obese mice, 2024, 0031-8655, 10.1111/php.14041 | |
604. | Michael R. Hamblin, 2024, Chapter 4, 978-3-031-74313-9, 53, 10.1007/978-3-031-74314-6_4 | |
605. | Marjorie Dole, Pierre Bleuet, Vincent Auboiroux, Malvina Billères, John Mitrofanis, Monte Carlo simulations of a multisource transcranial photobiomodulation helmet device: application to young and aged brains, 2024, 1, 3050-6816, 261, 10.4103/ATN.ATN-D-24-00022 | |
606. | Reiza Ventura, Jun‐Sang Bae, Eun Hee Kim, A. Young Kim, Min Hyuck Oh, Ji Hye Kim, Shin Hyuk Yoo, Gwanghui Ryu, Ji‐Hun Mo, Evaluating the Therapeutic Potential of Microneedle Patch Laser With Multiple Wavelengths in Allergic Rhinitis: Insights From an Allergic Rhinitis Mouse Model, 2024, 0196-8092, 10.1002/lsm.23862 | |
607. | Ruth Phypers, Venera Berisha-Muharremi, Reem Hanna, The Efficacy of Multiwavelength Red and Near-Infrared Transdermal Photobiomodulation Light Therapy in Enhancing Female Fertility Outcomes and Improving Reproductive Health: A Prospective Case Series with 9-Month Follow-Up, 2024, 13, 2077-0383, 7101, 10.3390/jcm13237101 | |
608. | Guillaume Blivet, François J. Roman, Julien Delrieu, Jacques Touchon, Translation from Preclinical Research to Clinical Trials: Brain–Gut Photobiomodulation Therapy for Alzheimer's Disease, 2024, 23, 0219-6352, 10.31083/j.jin2303057 | |
609. | Guillaume Blivet, François J. Roman, Benjamin Lelouvier, Céline Ribière, Jacques Touchon, Photobiomodulation Therapy: A Novel Therapeutic Approach to Alzheimer's Disease Made Possible by the Evidence of a Brain–Gut Interconnection, 2024, 23, 0219-6352, 10.31083/j.jin2305092 | |
610. | Laura Bernabei, Beniamino Leone, Daniele Hirsch, Valentina Mentuccia, Alessia Panzera, Francesco Riggio, Loredana Sangiovanni, Valentina Piserchia, Giuseppe Nicolò, Enrico Pompili, Neuromodulation Strategies in Lifelong Bipolar Disorder: A Narrative Review, 2024, 14, 2076-328X, 1176, 10.3390/bs14121176 | |
611. | Jan Martel, Nicolas Rouleau, Nirosha J. Murugan, Wei-Chun Chin, David M. Ojcius, John D. Young, Effects of light, electromagnetic fields and water on biological rhythms, 2024, 23194170, 100824, 10.1016/j.bj.2024.100824 | |
612. | Gisela Cristina Vianna Camolesi, Ahmed Samir El Kattan, José Lopez-Lopez, Andrés Blanco-Carrión, Abel García-García, Pilar Gándara-Vila, Mario Pérez-Sayáns, Pain, oedema and trismus responses following photobiomodulation therapy immediately after lower third molar extraction: results of a randomised, doble-blind and split mouth clinical trial, 2024, 15323382, 102080, 10.1016/j.jebdp.2024.102080 | |
613. | Mohd. Afzal Khan, Gehan Fatima, Akm Ashiquzzaman, Sang Seong Kim, Hyuksang Kwon, Young Ro Kim, Euiheon Chung, Evaluating the Preclinical Efficacy of Photobiomodulation in Alleviating Neuropathic Corneal Pain: A Behavioral Study, 2024, 26669145, 100680, 10.1016/j.xops.2024.100680 | |
614. | Changrui Zhao, Kun Fu, Jiameng Tian, Tian Long, Jianzhong Song, Siyu Chen, Chang Liu, Wearable photobiomodulation halts thyroid cancer growth by leveraging thyroid photosensitivity, 2024, 2380-6761, 10.1002/btm2.10734 | |
615. | Alexander R Guillen, Dennis Q Truong, Paula Cristina Faria, Brian Pryor, Luis De Taboada, Abhishek Datta, 2024, Computational analysis of light diffusion and thermal effects during Transcranial Photobiomodulation, 979-8-3503-7149-9, 1, 10.1109/EMBC53108.2024.10782579 | |
616. | Qiqi Fu, Jiali Yang, Hui Jiang, Yi Ren, Longfei Huo, Muqing Liu, Pulsed Red Photobiomodulation Boosts the Inhibition of Oxytocin‐Induced Primary Dysmenorrhea in Mice by Suppressing Oxidative Stress and Inflammation, 2024, 1864-063X, 10.1002/jbio.202400398 | |
617. | Maram Khaled, Adriana Baranov, Alvaro Diaz, Mansi Patel, Sarah Clements, Parsa Farsinejad, Kabir Khatana, Ashmitha Gnanapragasam, Sathurthika Selvanayagam, Zeineb Muhsen, Jocelyn Chan, Sanjum Hunjan, Ayman Kazi, Sapna Sharma, Lea Luketic, Joycelyne Efua Ewusie, Daniel Cordovani, Harsha Shanthanna, Michael R Hamblin, Photobiomodulation as part of multimodal analgesia to improve pain relief and wound healing after elective caesarean section: A protocol for randomized controlled trial, 2024, 19, 1932-6203, e0314010, 10.1371/journal.pone.0314010 | |
618. | Jaroslava Joniová, Aurélien Gregor, Martine Lambelet, Sébastien Déglise, Florent Allagnat, Georges Wagnières, Optimizing Photobiomodulation Radiometric and Spectral Parameters In Vitro to Enhance Angiogenesis and Mitochondrial Function, 2024, 26, 1422-0067, 93, 10.3390/ijms26010093 | |
619. | Aysin Erboz, Elif Kesekler, Pier Luigi Gentili, Vladimir N. Uversky, Orkid Coskuner-Weber, Electromagnetic Radiation and Biophoton Emission in Neuronal Communication and Neurodegenerative Diseases, 2024, 00796107, 10.1016/j.pbiomolbio.2024.12.004 | |
620. | Ehsan Hajesmaelzade, Mohammad Mohammadi, Sina Kakooei, Luca Solimei, Stefano Benedicenti, Nasim Chiniforush, Efficacy of Photobiomodulation Therapy Utilizing 808 nm and 660 nm Alone and in Combination for Treatment of Paresthesia in Rats, 2024, 13, 2227-9059, 65, 10.3390/biomedicines13010065 | |
621. | Ronald B. Koh, 2025, 9781394205325, 445, 10.1002/9781394205356.ch35 | |
622. | Régia Carla Medeiros da Silva, Lucas Gabriel Cunha da Silva, Natália Teixeira da Silva Cruz, Ádylla Rominne Lima Barbosa, Paulo Raphael Leite Maia, Ana Rafaela Luz de Aquino Martins, Evaluation of two low-level laser techniques as an adjunct to basic periodontal therapy: a randomized clinical trial, 2025, 40, 1435-604X, 10.1007/s10103-024-04275-w | |
623. | N. S. Yubitskaya, M. V. Antonyuk, N. D. Rozhkova, Laser therapy capabilities using Multiwave Locked System in pulmonology, 2024, 1998-5029, 158, 10.36604/1998-5029-2024-94-158-167 | |
624. | Chelsea Tripp, Lisa A. Miller, 2025, 9781394205325, 233, 10.1002/9781394205356.ch19 | |
625. | Max Meuser, Susanne Schwitzer, Parisa Faraji, Arne Ernst, Dietmar Basta, Peri-Traumatic Near-Infrared Light Treatment Attenuates the Severity of Noise-Induced Hearing Loss by Rescuing (Type I) Spiral Ganglion Neurons in Mice, 2025, 15, 2076-3425, 62, 10.3390/brainsci15010062 | |
626. | Jieun Jung, Tae Kim, Photobiomodulation and Its Therapeutic Potential in Sleep Disturbances, 2024, 15, 2093-9175, 218, 10.17241/smr.2024.02593 | |
627. | Yun Zhao, Na Zhang, Zhuowei Zhang, Ping Liu, Jianfei Dong, 2024, Efficacy of Three Red-NIR Wavelengths in Reducing Oxidative Stress Induced by Impaired Mitochondrial Functions, 979-8-3315-4114-9, 522, 10.1109/SSLCHINAIFWS64644.2024.10835359 | |
628. | Fatma Oner, Alpdogan Kantarci, Periodontal response to nonsurgical accelerated orthodontic tooth movement, 2025, 0906-6713, 10.1111/prd.12623 | |
629. | Patricia Brassolatti, Nivaldo Antonio Parizotto, Elaine Caldeira de Oliveira Guirro, Leandro Augusto de Almeida, Carla Roberta Tim, Michele Akemi Nishioka, José Ricardo de Souza, Ana Laura Martins de Andrade, Systemic photobiomodulation: an integrative review of evidence for intravascular laser irradiation of blood and vascular photobiomodulation, 2025, 40, 1435-604X, 10.1007/s10103-024-04233-6 | |
630. | Hadari Yotam, Pesah Galia, Goor-Aryeh Itay, Ungar Lior, Assessing the effectiveness of the SOLIO Alfa Cure Plus device in treating low back pain: a randomized controlled study, 2025, 20, 1749-799X, 10.1186/s13018-024-05441-0 | |
631. | Mario Álvarez-Martínez, Gabriel Borden, A systematic review on whole-body photobiomodulation for exercise performance and recovery, 2025, 40, 1435-604X, 10.1007/s10103-025-04318-w | |
632. | Andrew R. Stevens, Mohammed Hadis, Hannah Alldrit, Michael R. Milward, Valentina Di Pietro, Deena M. A. Gendoo, Antonio Belli, William Palin, David J. Davies, Zubair Ahmed, Evaluation of transcriptomic changes after photobiomodulation in spinal cord injury, 2025, 15, 2045-2322, 10.1038/s41598-025-87300-4 | |
633. | Yuli Fradkin, Joaquin A. Anguera, Alexander J. Simon, Luis De Taboada, Eugenia Steingold, Transcranial photobiomodulation for reducing symptoms of autism spectrum disorder and modulating brain electrophysiology in children aged 2–7: an open label study, 2025, 4, 2813-4540, 10.3389/frcha.2025.1477839 | |
634. | Thalita Molinos Campos, Mayra Costanti Vilela Campos, Raquel Agnelli Mesquita-Ferari, Anna Carolina Ratto Tempestini Horliana, Sandra Kalil Bussadori, Cinthya Cosme Gutierrez Duran, Alexandre Padilha, Aldo Brugnera Júnior, Samir Nammour, Ricardo Scarparo Navarro, Kristianne Porta Santos Fernandes, Lara Jansiski Motta, Cost-Effectiveness Analysis of Photobiomodulation After Third Molar Extraction for Pain Control, 2025, 22, 1660-4601, 159, 10.3390/ijerph22020159 | |
635. | S. F. Khattab, Y. F. Gomaa, E. A. E. Abdelaziz, N. M. A. Khattab, Influence of photobiomodulation therapy on regenerative potential of non-vital mature permanent teeth in healthy canine dogs, 2025, 1818-6300, 10.1007/s40368-025-01000-1 | |
636. | Sultan Albeshri, Raed AlRowis, Effect of mechanical debridement with and without adjunct antimicrobial photodynamic therapy for the treatment of peri-implant disease in obese patients: A systematic review and meta-analysis of randomized controlled trials, 2025, 52, 15721000, 104510, 10.1016/j.pdpdt.2025.104510 | |
637. | Yanjun Dan, Li Chen, Shanglin Jin, Jingtao Zhang, Yijian Zhu, Wenjuan Ma, Ziqi Liu, Jianfeng Luo, Chengfeng Zhang, Leihong Flora Xiang, The Therapeutic and Preventive Effects of Light‐Emitting Diode (LED) for Post‐Inflammatory Erythema and Hyperpigmentation: A Pilot Study, 2025, 41, 0905-4383, 10.1111/phpp.70003 | |
638. | Helya Helali, Nastaran Samani, Faraj Tabeie, Shiva Eiliaei, Ali Kheradmand, The effectiveness of Transcranial Photobiomodulation therapy (tPBM) on reducing anxiety, depression, and opioid craving in patients undergoing methadone maintenance treatment: a double-blind, randomized, controlled trial, 2025, 25, 1471-244X, 10.1186/s12888-025-06555-3 | |
639. | Mark Cronshaw, Steven Parker, Omar Hamadah, Josep Arnabat-Dominguez, Martin Grootveld, Photobiomodulation LED Devices for Home Use: Design, Function and Potential: A Pilot Study, 2025, 13, 2304-6767, 76, 10.3390/dj13020076 | |
640. | Chenghui Lu, Lanqing Mo, Xin Li, Guanhuan Du, Zhengquan Chen, Fan Wu, Lili Cai, Qing Du, Guoyao Tang, Effects of low-level light therapy on pain and related lesions in patients with oral lichen planus: A systematic review and meta-analysis, 2025, 15323382, 102126, 10.1016/j.jebdp.2025.102126 | |
641. | Vitor Fortuna, Gabriel F. Oliveira, Livia M. Xavier, Daniele V. Oliveira, Jaqueline G. Lima, Yasmin S. Oliveira, Beatriz S. Costa, Giselle B. Jesus, Setondji C.M.A. Yahouedehou, Elissandra M. Zanchin, José Roberto Meyer, José V. Meneses, Marilda Souza Gonçalves, Vanderlei S. Bagnato, Enhancing Sickle Cell Leg Ulcer Healing with Combined Photodynamic and Photobiomodulation Therapies: a pilot experience, 2025, 0965206X, 100879, 10.1016/j.jtv.2025.100879 | |
642. | 潮香 蒋, The Application of New Laser Technology in Gynecology, 2025, 15, 2161-8712, 116, 10.12677/acm.2025.153594 | |
643. | Patrick Xiang Ji, Lauren Pickel, Alan R. Berger, Nirojini Sivachandran, Improvement in Dry Age-Related Macular Degeneration with Photobiomodulation, 2025, 16, 1663-2699, 155, 10.1159/000543971 | |
644. | Parisa Ashrafi, Soyar Sari, Fatemeh Javani Jouni, Jaber Zafari, Fatemeh Asgari, Potentiated Effects of Photobiomodulation and Celecoxib on the Epithelial–Mesenchymal Transition Signaling of E-Cadherin, N-Cadherin, α-SMA in Breast Cancer Cells, MCF7, and MDA-MB-231, 2025, 2578-5478, 10.1089/photob.2024.0155 | |
645. | Davide Frumento, Ștefan Ţălu, Light-based technologies in immunotherapy: advances, mechanisms and applications, 2025, 17, 1750-743X, 123, 10.1080/1750743X.2025.2470111 | |
646. | Dimakatso B. Gumede, Nicolette N. Houreld, Photobiomodulation Promotes Wound Healing in a Diabetic Cellular Model, 2025, 2970, 1742-6588, 012014, 10.1088/1742-6596/2970/1/012014 | |
647. | Kai-Yang Chen, Hung Kuan Lee, Hoi-Chun Chan, Chi-Ming Chan, Is Multiwavelength Photobiomodulation Effective and Safe for Age-Related Macular Degeneration? A Systematic Review and Meta-Analysis, 2025, 2193-8245, 10.1007/s40123-025-01119-w | |
648. | Leonardo Díaz, Lukas Restelli, Emilia Valencia, Damla Ilhan Atalay, José Manuel Abarca, Alain Chalple Gil, Eduardo Fernández, Effectiveness of low-level laser therapy on temporomandibular disorders. A systematic review of randomized clinical trials., 2025, 15721000, 104558, 10.1016/j.pdpdt.2025.104558 | |
649. | E-Liisa Laakso, Tatjana Ewais, Katie McMahon, Josephine Forbes, Liza Phillips, Efficacy and Safety of Photobiomodulation in MELAS: Protocol for a Series of N-of-1 Trials, 2025, 14, 2077-0383, 2047, 10.3390/jcm14062047 | |
650. | Joanna Woźniak, Michał Pazdrak, Ada Domanasiewicz, Jakub Kaźmierski, Near-Infrared Stimulation in Psychiatry Disorders: A Systematic Review of Efficacy and Biological Mechanisms, 2025, 6, 2673-4087, 26, 10.3390/neurosci6010026 | |
651. | Hiro Kinugawa, Fumitaka Kawakami, Katsuhiko Miyamoto, Hiroshi Kumagai, Emiyu Ogawa, James D. Carroll, Ann Liebert, Jeri-Anne Lyons, 2025, In vitro study of optical vortex and photobiomodulation effects for the development of Parkinson’s disease treatment, 9781510683426, 9, 10.1117/12.3051919 | |
652. | Patricia Costa Oliveira, Luisa Oliveira Correia, Natalia Medeiros Dias Lopes, Gabriel Rodrigues Suassuna, Richard L. Doty, Fabio de Rezende Pinna, Richard Louis Voegels, Marco Aurelio Fornazieri, Efficacy of the adjunctive use of photobiomodulation therapy in olfactory disorders in post-COVID-19 patients: A randomized controlled trial, 2025, 91, 18088694, 101583, 10.1016/j.bjorl.2025.101583 | |
653. | Shota Sasaki, Hiroshi Kumagai, Shun Takeda, Tomohiro Kosuge, James D. Carroll, Ann Liebert, Jeri-Anne Lyons, 2025, Proposal for suitable classification of photomedical concepts, 9781510683426, 1, 10.1117/12.3040865 | |
654. | Sungkyoo Lim, Seihwan Choi, James D. Carroll, Ann Liebert, Jeri-Anne Lyons, 2025, Harnessing photobiomodulation therapy: a holistic approach to healthcare and wellness for the MZ generation, 9781510683426, 4, 10.1117/12.3048445 |