
Atopic dermatitis (AD) is a skin disease. In this study, we observed the effects of Nandina domestica (ND) on AD-induced NC/Nga mice with 1-chloro-2,4-dinitrobenzene (DNCB)-treatment. ND significantly ameliorated the AD-skin signs with reducing of skin thickness and mast cell infiltration in the injuried skin of DNCB-induced AD model. Furthermore, ND reduced serum immunoglobulin E (IgE) levels and restored into normal condition the total cell number (TCN) in dorsal skin tissue, the axillary lymph node (ALN), and spleen after DNCB exposure. ND also decreased the number of CD23+/B220+ cells in the ALN and CD3+ cells in dorsal skin. Moreover, it decreased the interleukin (IL)-4, (IL)-5 and IL-13 levels but increased the interferon-γ levels in splenocytes. Immunohistofluorescence staining showed that ND significantly increased claudin1 and Sirt1 protein expressions in the AD-induced mice. Our results suggested that ND significantly ameliorates DCNB-induced AD.
Citation: Mi-Young Yun, Kyoung-Sook Park, Kyung-Ah Yoon, Hwa-Jung Choi. Leaf extracts of Nandina domestica Thunb ameliorate atopic dermatitis in NC/Nga mice treated with 1-chloro-2,4-dinitrobenzene (DNCB)[J]. AIMS Bioengineering, 2021, 8(2): 154-164. doi: 10.3934/bioeng.2021014
[1] | Jennifer L. Wood, Wuxing Liu, Caixian Tang, Ashley E. Franks . Microorganisms in heavy metal bioremediation: strategies for applying microbial-community engineering to remediate soils. AIMS Bioengineering, 2016, 3(2): 211-229. doi: 10.3934/bioeng.2016.2.211 |
[2] | Kevyn Melo Lotas, Raissa Sayumy Kataki Fonseca, Joice Camila Martins da Costa, Ana Claudia Alves Cortez, Francisca das Chagas do Amaral Souza, Márcio Rodrigues Barreto, Lívia Melo Carneiro, João Paulo Alves Silva, Eveleise Samira Martins Canto, Flávia da Silva Fernandes, João Vicente Braga de Souza, Érica Simplício de Souza . Optimization of laccase production by Pleurotus ostreatus (Jacq.) P. Kumm. using agro-industrial residues: a comparative study on peels of tucumã (Astrocaryum aculeatum G. Mey.) and pupunha (Bactris gasipaes Kunth) fruits. AIMS Bioengineering, 2024, 11(4): 561-573. doi: 10.3934/bioeng.2024025 |
[3] | Nikolaos Giormezis, Konstantinos Papakonstantinou, Fevronia Kolonitsiou, Eleanna Drougka, Antigoni Foka, Styliani Sarrou, Evangelos D. Anastassiou, Efthimia Petinaki, Iris Spiliopoulou . Biofilm synthesis and its relationship with genetic characteristics in clinical methicillin-resistant staphylococci. AIMS Bioengineering, 2015, 2(4): 375-386. doi: 10.3934/bioeng.2015.4.375 |
[4] | Urooj Ainuddin, Maria Waqas . Finite state machine and Markovian equivalents of the lac Operon in E. coli bacterium. AIMS Bioengineering, 2022, 9(4): 400-419. doi: 10.3934/bioeng.2022029 |
[5] | Zongyuan Zhu, Rachael Simister, Susannah Bird, Simon J. McQueen-Mason, Leonardo D. Gomez, Duncan J. Macquarrie . Microwave assisted acid and alkali pretreatment of Miscanthus biomass for biorefineries. AIMS Bioengineering, 2015, 2(4): 449-468. doi: 10.3934/bioeng.2015.4.449 |
[6] | Guang Long, Bo Lin, Lu Wang, Lingyan Wu, Tieying Yin, Donghong Yu, Guixue Wang . Sappan Lignum Extract Inhibits Restenosis in the Injured Artery through the Deactivation of Nuclear Factor-κB. AIMS Bioengineering, 2014, 1(1): 25-39. doi: 10.3934/bioeng.2014.1.25 |
[7] | Shaoyung Chen, Hsinyi Mao, Pinhua Tu, Naichen Cheng, Jiashing Yu . Fabrication of decellularized adipose tissue/alginate composite microspheres with pASCs encapsulation for tissue engineering. AIMS Bioengineering, 2017, 4(3): 351-365. doi: 10.3934/bioeng.2017.3.351 |
[8] | Tutuka Dlume, Nonso E. Nnolim, Uchechukwu U. Nwodo . Exiguobacterium acetylicum transformed poultry feathers into amino acids through an extracellular secretion of keratinolytic enzymes. AIMS Bioengineering, 2024, 11(4): 489-505. doi: 10.3934/bioeng.2024022 |
[9] | Marianthi Logotheti, Eleftherios Pilalis, Nikolaos Venizelos, Fragiskos Kolisis, Aristotelis Chatziioannou . Development and validation of a skin fibroblast biomarker profile for schizophrenic patients. AIMS Bioengineering, 2016, 3(4): 552-565. doi: 10.3934/bioeng.2016.4.552 |
[10] | Daniele D’Arrigo, Marta Bottagisio, Silvia Lopa, Matteo Moretti, Arianna B. Lovati . Tissue engineering approaches to develop decellularized tendon matrices functionalized with progenitor cells cultured under undifferentiated and tenogenic conditions. AIMS Bioengineering, 2017, 4(4): 431-445. doi: 10.3934/bioeng.2017.4.431 |
Atopic dermatitis (AD) is a skin disease. In this study, we observed the effects of Nandina domestica (ND) on AD-induced NC/Nga mice with 1-chloro-2,4-dinitrobenzene (DNCB)-treatment. ND significantly ameliorated the AD-skin signs with reducing of skin thickness and mast cell infiltration in the injuried skin of DNCB-induced AD model. Furthermore, ND reduced serum immunoglobulin E (IgE) levels and restored into normal condition the total cell number (TCN) in dorsal skin tissue, the axillary lymph node (ALN), and spleen after DNCB exposure. ND also decreased the number of CD23+/B220+ cells in the ALN and CD3+ cells in dorsal skin. Moreover, it decreased the interleukin (IL)-4, (IL)-5 and IL-13 levels but increased the interferon-γ levels in splenocytes. Immunohistofluorescence staining showed that ND significantly increased claudin1 and Sirt1 protein expressions in the AD-induced mice. Our results suggested that ND significantly ameliorates DCNB-induced AD.
Atopic dermatitis (AD) is a atopic eczema showing inflammatory and intensely pruritic skin disease [1]. AD is raised by various interactions among environmental factors and disintergration of the immunity. AD showed various symptoms containing a fluctuating course, hyperactive immune response to environmental factors, skin barrier disintergration, drying, and itching [2].
AD exhibited skin thickening with characterstic infiltration of activated T cells and monocytes/macrophages within the derma [3]. Most of patients possessing AD show increased serum total IgE against various allergens [4]. Furethermore, the expression of T helper (Th) 2 cytokines (IL-4, IL-5, and IL-13) is increased in the AD-induced injured skin of AD [5], proposing that Th2 cells play important roles in disease progressing by AD.
In several reports, the pathogenesis and development of AD have been mostly attributed to the Th1/Th2 immune dysregulation. Accordingly, most therapies of AD have been conducted to reduce Th2-mediated immune responses [6]. However, raising many evidences have focused on a important role of structural abnormalities in the stratum corneum (SC) of AD pathophysiology [7]. Although any connection against the correlation of skin barrier disintergration and abnormal immune esponse still needs to be investigated, recent studies have reported the high synergism in dysregulation of immune response [8]. The reports show that both breaking skin barrier and abnormal immune responses are important factors in the progressing of AD and therefore, the multi-pronged treatment such as recovery of skin barriers and suppression of skin inflammation is essential for the effective method decreasing development of AD [9].
AD care is devoted on alleviating itching, repairing disintergration of skin barrier and attenuating the skin lesions. Most patients suffering from AD used emollients to improve health of skin and decrease itching of skin as well as anti-inflammatory agents such as glucocorticoids, cyclosporine A, azathioprine, and mycophenolate mofetil [10]. However, many patients limited current therapies becuase of their side effects [11]. Conventional products such as creams and ointments also were limited topical use on their skin. Therefore, passionate needs for novel topical agents generated to provide more effective treatment of AD skin lessions [12].
Nandina domestica (family of Berberidaceae) have its origin from China and Japan [13]. N. domestica has been used as a effective treatment to reduce dermatophytic infections [14]. Our previous study reported that N. domestica extracts possessing anti-oxidant property show anti-inflammatory effect by regulating mitogen activated protein kinases (MAPKs) signaling in lipopolysaccharide (LPS)-induced RAW264.7 cells [15]. In addition, N. domestica leaf extracts are known to possess anti-bacterial effect [16]. However, AD efficacy of the leaf extracts of N. domestica has not been adequately reported. Therefore, we explored the anti-atopic efficacy of leaf extracts of N. domestica and the related mechanisms.
Seven-week-old ALC NC/Nga Tnd mice were purchased from Orient (Sungnam, South Korea) and the mice were kept in specific pathogen free conditions. To start the animal studies, the mice were randomly divided into 4 groups including 5 mice per each group [no treatment (NC/Nga_Nr), dinitrochlorobenzene (DNCB) treatment (DNCB-CTL), dexamethasone plus DNCB (DNCB_Dexa), Nandina domestica Thunb extracts plus DNCB (DNCB_ND)]. All mice keeped in a cage without pathogen and provided ad libitum access to foods and waters. Our animal studies were conducted in accordance with relevant guidelines, and regulations and procedures related with mice were approved by the Institutional Animal Care and Use Committee (IACUC) of Daejeon University, republic of Korea (DJUARB2018-041).
To progress AD-skin lesions, the back skin of mice (week-8) were shaved. The mice were sensitized twice per week by applying 200 µL of 1% DNCB in acetone (3): olive oil (1). Seven days later, the second challenge was conducted with 150 µL of 0.4% DNCB three times per week for 5 weeks in order for the mice to retain the disease. The sensitized mice were classified into four groups: NC/Nga_Nr is normal control. DNCB_CTL is DNCB-treated control. DNCB_Dexa is 3 mg/kg dexamethasone. DNCB_ND is 300 mg/kg ND. Control mice were colored with acetone: olive oil (3:1) ND suspended in 0.5% carboxymethyl cellulose (CMC) was treated by oral administration daily for 5 weeks (300 mg/kg). Dexamethasone was used as a positive control.
Clinical observation to evaluate changes in the skin of ALC NC/Nga Tnd mice was performed at 1, 2 and 5-week for 5 weeks. The harshness of AD-skin lesions was examined by a previsously reported method based on skin symptoms such as scarring/dryness and excoriation/erosion symptoms [17]. Briefly, the skin clinical scoring combined an assessment of skin disease extrent with three clinical characters: erythema/hemorrhage, scarring/dryness and excoriation/erosion. The features are assessed on a representative area for a given intensity item, also on a scale of 0–3 (0 is none, 1 is mild, 2 is moderate and 3 is severe). The back skin of the mice were biopsied. After embedding by paraffin wax, the back skin cut in 4 µm thickness. The analysis of the AD-skin was conducted by hematoxylin and eosin (H&E) and toluidine blue (TB) staining. All samples were investigated under an inverted microscope (Nikon Eclipse Ti, Nikon, Tokyo, Japan).
Axillary lymph nodes (ALNs) and spleens were isolated from mice. After crushing, they were filtered by a cell strainer (70 µm). The filtered cells were conducted centrifugation at 3000× g for 5 min, primary spleen cells were put at ACK lysis buffer (Red Blood Cell Lysis, Thermo Scientific, Waltham, MA, USA) to elinimate red blood cells (RBC) for 5 min. They were washed twice with RPMI-1640 medium containing 10% FBS. After isolation of primary ALNs and spleen cells, they were resuspended in RPMI-1640 medium containing 10% FBS. Briefly, the dorsal skin was eliminated from the mice. After mincing with scalpels, it was incubated in PBS including 1 mg/ml collagenase IV (Sigma-C5138, Sigma, St. Louis, MO, USA) at 37 °C for 40 mins. The each samples were actively down to further separate the remaining tissue clumps by pipetting. The cell suspensions were filtered with a pore nylon cell strainer (BD Falcon, Bedford, MA, USA) of 70 mm and centrifuged for 20 min (450× g). The cell pellets were gathered. After washing twice, total cell number was collected with a hemocytometer chamber (Thermo Fisher Scientific, Grand Island, NY, USA). The flow cytometry analysis was conducted with suitable antibodies after staining of the cells.
Spleens from mice were excluded. To gain single-cell suspensions, the RBC were excluded using RBC lysis buffer. The isolated splenocytes were incubated with or without anti-CD3 antibody (0.5 µg/mL) (eBioscience, San Diego, CA, USA). After incubation for 24 h, the supernatant was gathered to decide the IL-4, IL-5, IL-13, and IFN-γ levels.
The minced dorsal skins of mice were incubated in PBS containing collagenase IV (1 mg/mL) and dispase (2 mg/mL) for 40 min at 37 °C. After staining the cells with nine antibodies (anti-CD3, anti-CD4, anti-CD8, anti-CD19, anti-CD23, anti-CD69, anti-B220, anti-CCR3 and anti-CD11b; BD Biosciences Pharmingen, San Diego, CA, USA) in staining buffer (PBS containing 1% v/v fetal bovine serum and 0.01% w/v sodium azide) for 30 min below ice, the cells were analyzed by a fluorescence-activated cell sorting analyzer with Cell-Quest software (BD Biosciences).
The IgE levels were assessed by ELISA kits in plasma, and IL-4, IL-5, IL-13 and IFN-γ levels (R&D Systems, St. Louis, MO, USA) were assessed by ELISA kits in the supernatant of cultured splenocytes. The ELISA kits were used with the manufacturer's protocols.
Data are dispalyed as the means ± standard errors (SEMs). One-way analysis of variance (ANOVA) and Duncan's test were analyzed with Prism 7.0 (GraphPad Software Inc., San Diego, CA, USA).
To assess the potential of the ND on AD-like skin lesion, skin clinical scores were obtained by the AD-skin features. As shown in Figure 2a, the skin clinical scores of DNCB-control (CTL) were significantly high after the treatment of DNCB and DNCB-Dexa mice showed significantly reduced. Interestingly, after week-5, skin clinical scores of DNCB-ND mice went into significantly reverse compared to the NDCB-CTL mice.
To evaluate the efficacy of ND on hypertrophy and granulocyte infiltration of AD skin, the images were obtained by microscopy after applying with ND (Figure 2b,d). The repeated treatment with DNCB was increased skin thickening of AD-mice. However, ND was able to retrieval dermal thickness to almost normal mice compared to that in the NDCB-CTL group, showing that it decreases the hyperkeratinization of the skin in AD-mice and improves the histological expression of skin lesions of AD-mice. As shown in Figure 2c,e, the dorsal skin stained with TB exhibited a decrease in the infiltration of mast cells in the ND-treated group. The mast cell count (MCC) in ND-treated group was significantly smaller than DNCB_CTL group. These findings indicate that ND has anti-allergic activity.
To evaluate whether ND exert their efficacy through a IgE, serum levels of total IgE were gathered. Treatment repeated with DNCB caused a remarkable increment of total IgE (Figure 2f). However, serum levels of total IgE in ND treated mice reduced significantly more than that of the DNCB_CTL. In the total IgE, ND showed almost equal potency as compared to that of DNCB-Dexa. These results indicate that ND may play a important role in suppression of IgE production during the progress of AD.
The total cell numbers in the ALNs, spleen, and dorsal skin tissue statistically increased by DNCB and decreased by ND (Table 1). ND also decreased the absolute numbers of CD23+/B220+ double-positive cells. The absolute numbers of CD3+/CD19+, CD4+/CD8+, and CD4+/CD69+ cells didn't change in ALNs and the spleen. Infiltration of inflammatory cells - was lower in the dorsal skin in the DNCB_ND group, although CCR3+/CD11b+ cell infiltration didn't suggest significant change. These results show that ND reduces allergic cell numbers
Cell Phenotypes | NC/Nga-Nr | DNCB_CTL | DNCB_Dexa | DNCB_ND | |
ALN | Total ALN cells (×104/mL) | 5.3 ± 1.6 | 184.0 ± 22.5### | 38.3 ± 12.2*** | 94.0 ± 9.0** |
CD3+/CD19+ (×105 Cells) | 0.69 ± 0.31 | 28.3 ± 5.15### | 5.21 ± 1.17*** | 27.46 ± 4.76 | |
CD4+/CD8+ (×105 Cells) | 0.69 ± 0.13 | 23.84 ± 0.27### | 6.27 ± 0.73*** | 21.60 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 0.59 ± 0.15 | 4.10 ± 1.72### | 2.70 ± 0.66** | 3.70 ± 1.11 | |
CD23+/B220+ (×105 Cells) | 0.97 ± 5.50 | 140.0 ± 12.72### | 27.4 ± 5.12*** | 50.0 ± 4.15** | |
Spleen | Total spleen cells (×104/mL) | 665.0 ± 99.1 | 1585.0 ± 231.3## | 810.0 ± 143.9** | 850.0 ± 83.5 |
CD3+/CD19+ (×105 Cells) | 269 ± 0.31 | 563 ± 115.2# | 321.0 ± 72.4 | 467.0 ± 32.4 | |
CD4+/CD8+ (×105 Cells) | 118 ± 0.13 | 284 ± 32.7# | 227 ± 73.0 | 160.0 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 7.72 ± 37.79 | 18.50 ± 7.49 | 12.57 ± 4.07 | 12.5 ± 3.67 | |
CD23+/B220+ (×105 Cells) | 373.25 ± 86.49 | 616.21 ± 331.61# | 448.81 ± 65.24 | 575 ± 57.35 | |
Dorsal skin | Total dorsal skin cells (×104/mL) | 3.50 ± 1.10 | 27.5 ± 2.5### | 9.0 ± 1.0*** | 9.5 ± 1.5** |
CD3+(×105 Cells) | 0.30 ± 4.46 | 3.20 ± 0.31### | 1.80 ± 0.46** | 2.2 ± 0.16* | |
CCR3+/CD11b+ (×105 Cells) | 0.89 ± 0.06 | 2.86 ± 0.59## | 2.07 ± 0.03 | 1.26 ± 0.10 |
*Note: Each value represents mean ± SEM values for five mice. # p < 0.05, ## p < 0.01, and ### p < 0.001 compared with normal group (NC/Nga_Nr); * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with only DNCB group (DNCB-CTL).
We investigated whether ND may regulate the cytokines secreted by of Th1/Th2 cells in the AD mice. As shown in Figure 3, ND reduced the IL-4, IL-5 and IL-13 levels in cultured splenocytes. The level of IFN-γ increased compared to those in the NC/Nga_Nr. These results indicate that ND can improve AD-producted cytokines by regulating Th1 and Th2 cytokines.
Topical application with DNCB on the skin of mice significantly reduced expression of claudin 1 and sirt 1 proteins due to damage of the skin barrier. DNCB-ND significantly increased expression of claudin 1 and sirt 1 proteins. A weaker increment of two proteins was appeard in the DNCB-Dexa compared to the DNCB-ND (Figure 4). These results indicate that ND alleviates disruption of claudin 1 and sirt 1 proteins related with skin barrier.
AD is characterized by exacerbations and remissions of eczematous skin [18]. AD showed a chronic skin disorder with an interplay of migrating lymphocytes and epidermal keratinocytes (KC) [19]. AD-induced skin lesions are appeard dermal mononuclear infiltration and spongiosis in the epidermis of skin [20]. In this study, we examined whether ND could improve AD symptoms such as skin lesion and infiltration of mast cells. ND exhibited efficacy on improvement of skin clinical score and on obstruction of tissue mast cells infiltration with ameliorating the hyperkeratosis or thickening of the stratum corneum.
AD is generally showed increased allergen-specific IgE, mast cell activation and Th2 cytokine production in dermal lesions after continuous exposures to a special allergen [21]. Furthermore, IgE as a unique antibody possessed ability increasing the production of Th2-type cytokines as well as activation of mast cells, pointing its importance in makting a evil circle inducing to a chronic status in AD-patients [22]. In our study, ND showed inhibition of IgE and Th2 cytokines (IL-4, IL-5 and IL-13) levels but increment of Th1 cytokine (INF-γ). Furthermore, consistent with the reduced mast cell infiltration, ND potently ameliorated the levels of serum IgE and modulated Th2 cytokines and Th1 cytokine.
AD displayed accumulation of activated T cells in the skin and increased IgE production [23]. AD is generally characterized by increased numbers of infiltrating T cells and activated CD4+ or CD8+ T cells in the dermis. These lymphocytes are involved in the promotion of cell-mediated and humoral immunity through the secretion of various cytokines [24]. The problem, however, occurs when cell numbers are elevated because of the continuous presence of AD-inducing agents. At a previous study, double positive (DP) T cells expressing both CD4 and CD8 have been described in several pathological conditions as well as in normal individuals and a subset of DP T cells expressing high levels of both CD4 and CD8ab heterodimer (CD4hiCD8hi), has been identified in autoimmune and chronic inflammatory disorders [25]. The main finding of this study show that ND reduced the levels of CD3+ cells infiltration in the skin dorsal and lowered the numbers of CD4+/CD8+ cells in the ALNs and spleens. Simultaneously, ND restored the levels of IFN-γ secretion by splenocytes, as in the NC/Nga_Nr. However, ND lowered the levels of IL-4, IL-5 and IL-13 secreted by splenocytes.
Tight junctions (TJs) were involved in a cell-to-cell adhesion in the cells forming the epithelium and endothelium [26]. The TJ barrier is displayed below the stratum corneum barrier, and its constitutive proteins (occludin, claudin-1, claudin-4, and ZO-1) are displayed by colocalization in the granular layers [27].
Sirtuin 1 (Sirt1), which is an NAD-dependent protein deacetylase, relate with cell survival, metabolism, senescence, and stress response in several cells and it conduct a important role in tumorigenesis and homeostasis of skin [28],[29]. The claudin1 and sirt1 related abnormalities of barrier in the development of AD. In this study, ND increased protein expression levels of claudin 1 and Sirt1 decreased by AD in dorsal skin tissue, indicating that ND may ameliorate skin barrier functioning in AD.
ND improved AD symptoms and it reduced the infiltration of mast cells, the levels of inflammatory cells infiltration in the skin dorsal, the numbers of CD4+/CD8+ cells in the ALNs and spleens, the levels of IgE and Th2 cytokines in AD-mice. It also improved skin barrier functioning with increasing of claudin 1 and Sirt1 protein expression levels. Therefore, this report suggest that ND significantly ameliorates DCNB-induced AD.
[1] |
Misery L, Huet F, Gouin O, et al. (2019) Current pharmaceutical developments in atopic dermatitis. Curr Opin Pharmacol 46: 7-13. doi: 10.1016/j.coph.2018.12.003
![]() |
[2] |
Brunner PM (2019) Early immunologic changes during the onset of atopic dermatitis. Ann Allergy Asthma Immunol 123: 152-157. doi: 10.1016/j.anai.2019.03.033
![]() |
[3] |
Bieber T (2008) Atopic dermatitis. N Engl J Med 358: 1483-1494. doi: 10.1056/NEJMra074081
![]() |
[4] |
Ponyai G, Hidvegi B, Nemeth I, et al. (2008) Contact and aeroallergens in adulthood atopic dermatitis. J Eur Acad Dermatol Venereol 22: 1346-1355. doi: 10.1111/j.1468-3083.2008.02886.x
![]() |
[5] |
Hamid Q, Boguniewicz M, Leung DY (1994) Differential in situ cytokine gene expression in acute versus chronic atopic dermatitis. J Clin Invest 94: 870-876. doi: 10.1172/JCI117408
![]() |
[6] |
Leung DY (2000) Atopic dermatitis: New insights and opportunities for therapeutic intervention. J Allergy Clin Immunol 105: 860-876. doi: 10.1067/mai.2000.106484
![]() |
[7] |
Elias PM, Hatano Y, Williams ML (2008) Basis for the barrier abnormality in atopic dermatitis: Outside-inside-outside pathogenic mechanisms. J Allergy Clin Immunol 121: 1337-1343. doi: 10.1016/j.jaci.2008.01.022
![]() |
[8] |
Boguniewicz M, Leung DYM (2011) Atopic dermatitis: A disease of altered skin barrier and immune dysregulation. Immunol Rev 242: 233-246. doi: 10.1111/j.1600-065X.2011.01027.x
![]() |
[9] |
Shin KO, Ha DH, Kim JO, et al. (2020) Exosomes from human adipose tissue-derived mesenchymal stem cells promote epidermal barrier repair by inducing de novo synthesis of ceramides in atopic dermatitis. Cells 9: 680. doi: 10.3390/cells9030680
![]() |
[10] |
Schakel K, Dobel T, Bosselmann I (2014) Future treatment options for atopic dermatitis—small molecules and beyond. J Dermatol Sci 73: 91-100. doi: 10.1016/j.jdermsci.2013.11.009
![]() |
[11] |
Boguniewicz M, Alexis AF, Beck LA, et al. (2017) Expert perspectives on management of moderate-to-severe atopic dermatitis: A multidisciplinary consensus addressing current and emerging therapies. J Allergy Clin Immunol Pract 5: 1519-1531. doi: 10.1016/j.jaip.2017.08.005
![]() |
[12] | Akhtar N, Verma A, Pathak K (2017) Exploring preclinical and clinical effectiveness of nanoformulations in the treatment of atopic dermatitis: Safety aspects and patent reviews. Bull Fac Pharm Cairo Univ 55: 1-10. |
[13] | Guo ZY, Zhang ZY, Xiao JQ, et al. (2018) Antibacterial effects of leaf extract of Nandina domestica and the underlined nechanism. Evid Based Complement Alternat Med 2018: 8298151. |
[14] |
Bajpai VK, Yoon JI, Kang SC (2009) Antifungal potential of essential oil and various organic extracts of Nandina domestica Thunb. against skin infectious fungal pathogens. Appl Microbiol Biotechnol 83: 1127-1133. doi: 10.1007/s00253-009-2017-5
![]() |
[15] | Yun MY, Zhe W, Choi HJ (2020) Anti-inflammatory effect of Nandina domestica Thunb extracts possessing anti-oxidant property by regulating MAPKs signaling in LPS-induced inflammatory cells. J Altern Complement Integr Med 6: 114. |
[16] | Guo ZY, Zhang ZY, Xiao JQ, et al. (2018) Antibacterial effects of leaf extract of Nandina domestica and the underlined mechanism. Evid Based Complement Alternat Med 2018: 8298151. |
[17] |
Cabanillas B, Brehler AC, Novak N (2017) Atopic dermatitis phenotypes and the need for personalized medicin. Curr Opin Allergy Clin Immunol 17: 309-315. doi: 10.1097/ACI.0000000000000376
![]() |
[18] |
Akdis CA, Blaser K, Akdis M (2004) Apoptosis in tissue inflammation and allergic disease. Curr Opin Immunol 16: 717-723. doi: 10.1016/j.coi.2004.09.004
![]() |
[19] |
Verhagen J, Akdis M, Traidl-Hoffmann C, et al. (2006) Absence of T-regulatory cell expression and function in atopic dermatitis skin. J Allergy Clin Immunol 117: 176-183. doi: 10.1016/j.jaci.2005.10.040
![]() |
[20] |
Spergel JM, Mizoguchi E, Oettgen H, et al. (1999) Roles of TH1 and TH2 cytokines in a murine model of allergic dermatitis. J Clin Invest 103: 1103-1111. doi: 10.1172/JCI5669
![]() |
[21] |
Yoshimoto T, Yasuda K, Tanaka H, et al. (2009) Basophils contribute to TH2-IgE responses in vivo via IL-4 production and presentation of peptide–MHC class II complexes to CD4+ T cells. Nat Immunol 10: 706-712. doi: 10.1038/ni.1737
![]() |
[22] |
Leung DY, Bieber T (2003) Atopic dermatitis. Lancet 361: 151-160. doi: 10.1016/S0140-6736(03)12193-9
![]() |
[23] |
Liu FT, Goodarzi H, Chen HY (2011) IgE, mast cells, and eosinophils in atopic dermatitis. Clin Rev Allergy Immunol 41: 298-310. doi: 10.1007/s12016-011-8252-4
![]() |
[24] |
Ellingsen AR, Sorensen FB, Larsen JO, et al. (2001) Stereological quantification of lymphocytes in skin biopsies from atopic dermatitis patients. Acta Derm Venereol 81: 258-262. doi: 10.1080/00015550152572886
![]() |
[25] |
Parel Y, Chizzolini C (2004) CD4+ CD8+ double positive (DP) T cells in health and disease. Autoimmun Rev 3: 215-220. doi: 10.1016/j.autrev.2003.09.001
![]() |
[26] |
Tsukita S, Furuse M, Itoh M (2001) Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2: 285-293. doi: 10.1038/35067088
![]() |
[27] |
Brandner JM, Kief S, Grund C, et al. (2002) Organization and formation of the tight junction system in human epidermis and cultured keratinocytes. Eur J Cell Biol 81: 253-263. doi: 10.1078/0171-9335-00244
![]() |
[28] |
Haigis MC, Guarente LP (2006) Mammalian sirtuins—emerging roles in physiology, aging, and calorie restriction. Gene Dev 20: 2913-2921. doi: 10.1101/gad.1467506
![]() |
[29] |
Ming M, Shea CR, Guo X, et al. (2010) Regulation of global genome nucleotide excision repair by SIRT1 through xeroderma pigmentosum C. Proc Natl Acad Sci USA 107: 22623-22628. doi: 10.1073/pnas.1010377108
![]() |
1. | Huiqin Qian, Yanling Li, Nandina domestica Thunb.: a review of traditional uses, phytochemistry, pharmacology, and toxicology, 2024, 15, 1663-9812, 10.3389/fphar.2024.1407140 | |
2. | Hanna Koval, Mykola Bondarovych, Anatoliy Goltsev, Cord Blood as a Corrector of Hematological and Cytomorphological Characteristics of Lymphohemopoietic Organs in Experimental Atopic Dermatitis, 2023, 7, 2616-177X, 3, 10.20535/ibb.2023.7.3.280873 |
Cell Phenotypes | NC/Nga-Nr | DNCB_CTL | DNCB_Dexa | DNCB_ND | |
ALN | Total ALN cells (×104/mL) | 5.3 ± 1.6 | 184.0 ± 22.5### | 38.3 ± 12.2*** | 94.0 ± 9.0** |
CD3+/CD19+ (×105 Cells) | 0.69 ± 0.31 | 28.3 ± 5.15### | 5.21 ± 1.17*** | 27.46 ± 4.76 | |
CD4+/CD8+ (×105 Cells) | 0.69 ± 0.13 | 23.84 ± 0.27### | 6.27 ± 0.73*** | 21.60 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 0.59 ± 0.15 | 4.10 ± 1.72### | 2.70 ± 0.66** | 3.70 ± 1.11 | |
CD23+/B220+ (×105 Cells) | 0.97 ± 5.50 | 140.0 ± 12.72### | 27.4 ± 5.12*** | 50.0 ± 4.15** | |
Spleen | Total spleen cells (×104/mL) | 665.0 ± 99.1 | 1585.0 ± 231.3## | 810.0 ± 143.9** | 850.0 ± 83.5 |
CD3+/CD19+ (×105 Cells) | 269 ± 0.31 | 563 ± 115.2# | 321.0 ± 72.4 | 467.0 ± 32.4 | |
CD4+/CD8+ (×105 Cells) | 118 ± 0.13 | 284 ± 32.7# | 227 ± 73.0 | 160.0 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 7.72 ± 37.79 | 18.50 ± 7.49 | 12.57 ± 4.07 | 12.5 ± 3.67 | |
CD23+/B220+ (×105 Cells) | 373.25 ± 86.49 | 616.21 ± 331.61# | 448.81 ± 65.24 | 575 ± 57.35 | |
Dorsal skin | Total dorsal skin cells (×104/mL) | 3.50 ± 1.10 | 27.5 ± 2.5### | 9.0 ± 1.0*** | 9.5 ± 1.5** |
CD3+(×105 Cells) | 0.30 ± 4.46 | 3.20 ± 0.31### | 1.80 ± 0.46** | 2.2 ± 0.16* | |
CCR3+/CD11b+ (×105 Cells) | 0.89 ± 0.06 | 2.86 ± 0.59## | 2.07 ± 0.03 | 1.26 ± 0.10 |
*Note: Each value represents mean ± SEM values for five mice. # p < 0.05, ## p < 0.01, and ### p < 0.001 compared with normal group (NC/Nga_Nr); * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with only DNCB group (DNCB-CTL).
Cell Phenotypes | NC/Nga-Nr | DNCB_CTL | DNCB_Dexa | DNCB_ND | |
ALN | Total ALN cells (×104/mL) | 5.3 ± 1.6 | 184.0 ± 22.5### | 38.3 ± 12.2*** | 94.0 ± 9.0** |
CD3+/CD19+ (×105 Cells) | 0.69 ± 0.31 | 28.3 ± 5.15### | 5.21 ± 1.17*** | 27.46 ± 4.76 | |
CD4+/CD8+ (×105 Cells) | 0.69 ± 0.13 | 23.84 ± 0.27### | 6.27 ± 0.73*** | 21.60 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 0.59 ± 0.15 | 4.10 ± 1.72### | 2.70 ± 0.66** | 3.70 ± 1.11 | |
CD23+/B220+ (×105 Cells) | 0.97 ± 5.50 | 140.0 ± 12.72### | 27.4 ± 5.12*** | 50.0 ± 4.15** | |
Spleen | Total spleen cells (×104/mL) | 665.0 ± 99.1 | 1585.0 ± 231.3## | 810.0 ± 143.9** | 850.0 ± 83.5 |
CD3+/CD19+ (×105 Cells) | 269 ± 0.31 | 563 ± 115.2# | 321.0 ± 72.4 | 467.0 ± 32.4 | |
CD4+/CD8+ (×105 Cells) | 118 ± 0.13 | 284 ± 32.7# | 227 ± 73.0 | 160.0 ± 0.50 | |
CD4+/CD69+ (×105 Cells) | 7.72 ± 37.79 | 18.50 ± 7.49 | 12.57 ± 4.07 | 12.5 ± 3.67 | |
CD23+/B220+ (×105 Cells) | 373.25 ± 86.49 | 616.21 ± 331.61# | 448.81 ± 65.24 | 575 ± 57.35 | |
Dorsal skin | Total dorsal skin cells (×104/mL) | 3.50 ± 1.10 | 27.5 ± 2.5### | 9.0 ± 1.0*** | 9.5 ± 1.5** |
CD3+(×105 Cells) | 0.30 ± 4.46 | 3.20 ± 0.31### | 1.80 ± 0.46** | 2.2 ± 0.16* | |
CCR3+/CD11b+ (×105 Cells) | 0.89 ± 0.06 | 2.86 ± 0.59## | 2.07 ± 0.03 | 1.26 ± 0.10 |