Loading [MathJax]/jax/element/mml/optable/MathOperators.js
Review Special Issues

Molecular mechanisms in Alzheimer's disease and the impact of physical exercise with advancements in therapeutic approaches

  • Alzheimer's disease (AD) is one of the most common, severe neurodegenerative brain disorder characterized by the accumulation of amyloid-beta plaques, neurofibrillary tangles in the brain causing neural disintegration, synaptic dysfunction, and neuronal death leading to dementia. Although many US-FDA-approved drugs like Donepezil, Rivastigmine, Galantamine are available in the market, their consumption reduces only the symptoms of the disease but fails in potency to cure the disease. This disease affects many individuals with aging. Combating the disease tends to be very expensive. This review focuses on biochemical mechanisms in the neuron both at normal and AD state with relevance to the tau hypothesis, amyloid hypothesis, the risk factors influencing dementia, oxidative stress, and neuroinflammation altogether integrated with neurodegeneration. A brief survey is carried out on available biomarkers in the diagnosis of the disease, drugs used for the treatment, and the challenges in approaching therapeutic targets in inhibiting the disease pathologies. This review conjointly assesses the demerits with the inefficiency of drugs to reach targets, their side effects, and toxicity. Optimistically, this review directs on the advantageous strategies in using nanotechnology-based drug delivery systems to cross the blood-brain barrier for improving the efficacy of drugs combined with a novel neuronal stem cell therapy approach. Determinately, this review aims at the natural, non-therapeutic healing impact of physical exercise on different model organisms and the effect of safe neuromodulation treatments using repetitive Transcranial Magnetic Stimulation (rTMS), transcranial Electrical Stimulation (tES) in humans to control the disease pathologies prominent in enhancing the synaptic function.

    Citation: Kiran Kumar Siddappaji, Shubha Gopal. Molecular mechanisms in Alzheimer's disease and the impact of physical exercise with advancements in therapeutic approaches[J]. AIMS Neuroscience, 2021, 8(3): 357-389. doi: 10.3934/Neuroscience.2021020

    Related Papers:

    [1] Rina Su, Chunrui Zhang . The generation mechanism of Turing-pattern in a Tree-grass competition model with cross diffusion and time delay. Mathematical Biosciences and Engineering, 2022, 19(12): 12073-12103. doi: 10.3934/mbe.2022562
    [2] Yue Xing, Weihua Jiang, Xun Cao . Multi-stable and spatiotemporal staggered patterns in a predator-prey model with predator-taxis and delay. Mathematical Biosciences and Engineering, 2023, 20(10): 18413-18444. doi: 10.3934/mbe.2023818
    [3] Mingzhu Qu, Chunrui Zhang, Xingjian Wang . Analysis of dynamic properties on forest restoration-population pressure model. Mathematical Biosciences and Engineering, 2020, 17(4): 3567-3581. doi: 10.3934/mbe.2020201
    [4] Fang Liu, Yanfei Du . Spatiotemporal dynamics of a diffusive predator-prey model with delay and Allee effect in predator. Mathematical Biosciences and Engineering, 2023, 20(11): 19372-19400. doi: 10.3934/mbe.2023857
    [5] Ranjit Kumar Upadhyay, Swati Mishra . Population dynamic consequences of fearful prey in a spatiotemporal predator-prey system. Mathematical Biosciences and Engineering, 2019, 16(1): 338-372. doi: 10.3934/mbe.2019017
    [6] Sourav Kumar Sasmal, Jeet Banerjee, Yasuhiro Takeuchi . Dynamics and spatio-temporal patterns in a prey–predator system with aposematic prey. Mathematical Biosciences and Engineering, 2019, 16(5): 3864-3884. doi: 10.3934/mbe.2019191
    [7] Swadesh Pal, Malay Banerjee, Vitaly Volpert . Spatio-temporal Bazykin’s model with space-time nonlocality. Mathematical Biosciences and Engineering, 2020, 17(5): 4801-4824. doi: 10.3934/mbe.2020262
    [8] Meiling Zhu, Huijun Xu . Dynamics of a delayed reaction-diffusion predator-prey model with the effect of the toxins. Mathematical Biosciences and Engineering, 2023, 20(4): 6894-6911. doi: 10.3934/mbe.2023297
    [9] Tingting Ma, Xinzhu Meng . Global analysis and Hopf-bifurcation in a cross-diffusion prey-predator system with fear effect and predator cannibalism. Mathematical Biosciences and Engineering, 2022, 19(6): 6040-6071. doi: 10.3934/mbe.2022282
    [10] Xiaomei Bao, Canrong Tian . Turing patterns in a networked vegetation model. Mathematical Biosciences and Engineering, 2024, 21(11): 7601-7620. doi: 10.3934/mbe.2024334
  • Alzheimer's disease (AD) is one of the most common, severe neurodegenerative brain disorder characterized by the accumulation of amyloid-beta plaques, neurofibrillary tangles in the brain causing neural disintegration, synaptic dysfunction, and neuronal death leading to dementia. Although many US-FDA-approved drugs like Donepezil, Rivastigmine, Galantamine are available in the market, their consumption reduces only the symptoms of the disease but fails in potency to cure the disease. This disease affects many individuals with aging. Combating the disease tends to be very expensive. This review focuses on biochemical mechanisms in the neuron both at normal and AD state with relevance to the tau hypothesis, amyloid hypothesis, the risk factors influencing dementia, oxidative stress, and neuroinflammation altogether integrated with neurodegeneration. A brief survey is carried out on available biomarkers in the diagnosis of the disease, drugs used for the treatment, and the challenges in approaching therapeutic targets in inhibiting the disease pathologies. This review conjointly assesses the demerits with the inefficiency of drugs to reach targets, their side effects, and toxicity. Optimistically, this review directs on the advantageous strategies in using nanotechnology-based drug delivery systems to cross the blood-brain barrier for improving the efficacy of drugs combined with a novel neuronal stem cell therapy approach. Determinately, this review aims at the natural, non-therapeutic healing impact of physical exercise on different model organisms and the effect of safe neuromodulation treatments using repetitive Transcranial Magnetic Stimulation (rTMS), transcranial Electrical Stimulation (tES) in humans to control the disease pathologies prominent in enhancing the synaptic function.


    Abbreviations

    AD:

    Alzheimer's disease; 

    Aβ:

    Amyloid-beta; 

    NFTs:

    Neurofibrillary Tangles; 

    APP:

    Amyloid Precursor Protein; 

    sAPPα:

    soluble Amyloid Precursor Protein alpha; 

    AICD:

    Amyloid Precursor Protein Intra Cellular Domain; 

    NMDAR:

    N-Methyl-D-Aspartate Receptor; 

    AMPAR:

    α-Amino-3-hydroxy-5-Methyl-4-isoxazole Propionic acid Receptor; 

    nAChRs:

    nicotinic Acetylcholine Receptors; 

    mAChRs:

    muscarinic Acetylcholine Receptors; 

    sAPPβ:

    soluble Amyloid Precursor Protein beta; 

    ROS:

    Reactive Oxygen Species; 

    PKC:

    Protein Kinase C; 

    PKA:

    Protein Kinase A; 

    ERK2:

    Extracellular signal-Regulated Kinases2; 

    CDK5:

    Cyclin-Dependent Kinases 5; 

    AchE:

    AcetylcholinEsterase; 

    CSF:

    Cerebro Spinal Fluid; 

    PET:

    Positron Emission Tomography; 

    BBB:

    Blood-Brain Barrier; 

    PE:

    Physical Exercise; 

    DLB:

    Dementia with Lewy Bodies; 

    FTD:

    FrontoTemporal Dementia; 

    MRI:

    Magnetic Resonance Imaging; 

    fMRI:

    functional Magnetic Resonance Imaging; 

    rTMS:

    repetitive Transcranial Magnetic Stimulation; 

    tES:

    transcranial Electrical Stimulation; 

    tDCS:

    transcranial Direct Current Stimulation; 

    tACS:

    transcranial Alternating Current Stimulation; 

    tRNS:

    transcranial Random Noise Stimulation; 

    tPCS:

    transcranial Pulsed Current Stimulation; 

    MCI:

    Mild Cognitive Impairment; 

    Drp1:

    Dynamin-related protein-1; 

    PSEN 1:

    Presenilin-1; 

    PSEN 2:

    Presenilin-2; 

    IDE:

    Insulin Degrading Enzyme; 

    PD:

    Parkinson's Disease; 

    ApoE:

    Apolipoprotein-E; 

    PKB:

    Protein Kinase B; 

    GSK3α/β:

    Glycogen synthase kinase 3α/β; 

    ETC:

    Electron Transport Chain; 

    IGF-1:

    Insulin-like Growth Factor-1; 

    IL-6:

    Interleukin-6; 

    NOS-2:

    Nitrous Oxide Synthase-2; 

    NF-κB:

    Nuclear Factor Kappa B; 

    LRP-1:

    Lipoprotein Receptor-related Protein-1; 

    OR-A:

    Orexin-A; 

    OR-B:

    Orexin-B; 

    GPCRs:

    G-Protein-Coupled Receptors; 

    OX1R:

    Orexin Receptor-type-1; 

    OX2R:

    Orexin Receptor-type-2; 

    BDNF:

    Brain-Derived Neurotrophic Factor; 

    OSA:

    Obstructive Sleep Apnea (OSA); 

    CDH:

    Central Disorders of Hypersomnolence; 

    REM:

    Rapid Eye Movement; 

    EEG:

    Electroencephalogram; 

    TBI:

    Traumatic Brain Injury; 

    123I-MIBG:

    123I-Meta-IodoBenzylGuanidine; 

    [11C]PiB:

    [11C]-Pittsburgh compound B; 

    18F:

    Fluorine 18-labeled; 

    MoCA:

    Montreal Cognitive Assessment; 

    T-tau:

    Total tau; 

    P-tau:

    phosphorylated-tau; 

    VLP-1:

    Visinin-Like Protein; 

    NFL:

    Neuro Filament Light Protein; 

    HFABP:

    Heart Fatty Acid Binding Protein; 

    NSE:

    Neuron-Specific Enolase; 

    CHI3L1:

    Chitinase-3-like protein-1; 

    TNFSF10:

    Tumor Necrosis Factor Ligand Superfamily member-10; 

    ACE:

    Angiotensin-Converting Enzyme; 

    3xTg-AD:

    triple-transgenic-AD; 

    VBM:

    Voxel-Based Morphometry

    Mosquito-borne infectious diseases, a common type of vector-borne infections diseases, are primarily caused by pathogens, which are transmitted from mosquitoes to humans or other animals [1]. Mosquitoes are one of the most widely distributed vectors in the world, transmitting a variety of parasites and viruses. While the vector organisms may not develop the disease themselves, they serve as a means for the pathogen to spread among hosts. With mosquitoes found in various locations from tropical to temperate zones, mosquito-borne infections have a wide and diverse geographic distribution [2]. Diseases such as malaria, lymphatic filariasis, West Nile Virus, Zika virus, and dengue fever are commonly transmitted by mosquitoes, with no specific vaccine or medication available for treatment. These diseases pose a significant threat to human health and socio-economic development worldwide.

    Malaria is an infectious disease caused by a protozoan parasite that is mainly transmitted to humans through the bite of a mosquito [3]. The disease is widespread in tropical and subtropical regions, particularly in poorer areas of Africa, Asia, and Latin America. Globally, malaria remains a major public health problem, resulting in millions of infections and hundreds of thousands of deaths annually. Apart from malaria, as the classic mosquito-borne infectious disease, dengue fever also attracts considerable attention. Dengue fever is a serious infectious disease caused by the dengue virus, primarily transmitted to humans by the Aedes aegypti mosquito, but also through blood transfusion, organ transplantation, and vertical transmission [4]. In addition, both yellow fever and West Nile disease are also caused by mosquitoes carrying the corresponding viral infections [5,6]. Yellow fever is a rare disease among U.S. travelers. Conversely, West Nile virus is the primary mosquito-borne disease in the continental U.S., commonly transmitted through the bite of an infected mosquito.

    Research on mosquito-borne diseases has proliferated [7,8,9,10]. Mathematical models have been increasingly used for experimental and observational studies of different biological phenomena, and a wide range of techniques and applications have been developed to study epidemic diseases. For example, Newton and Reiter [8] developed a deterministic Susceptibility, Exposure, Infection, Resistance, and Removal (SEIR) model of dengue transmission to explore the behavior of epidemics and realistically reproduce epidemic transmission in immunologically unmade populations. Moreover, Pandey et al. [9] proposed a Caputo fractional order derivative mathematical model of dengue disease to study the transmission dynamics of the disease and to make reliable conclusions about the behavior of dengue epidemics. In addition, in order to investigate the effect of the vector on the dynamics of the disease, Shi and Zhao et al. [10] proposed a differential system with saturated incidence to model a vector-borne disease

    {˙SH=μK+dIHμSH(¯β1IV1+η1IV+¯β2IH1+η2IH)SH,˙IH=(¯β1IV1+η1IV+¯β2IH1+η2IH)SH(d+μ+γ)IH,˙RH=γIHμRH,˙SV=Λβ3IHSV1+η3IHmSV,˙IV=β3IHSV1+η3IHmIV, (1.1)

    the biological significance of the model (1.1) parameters are shown in Table 1. In this model, there are two populations, namely mosquito vector population and human host population. The mosquito vector population is divided into two categories, SV and IV, and N=SV+IV, while human host population is divided into three categories, SH,IH and RH. According to [10], it is reasonable to assume that the total number of human K=SH+IH+RH is a positive constant.

    Table 1.  Variables and parameters in model (1.1).
    Variables and Parameters Description
    SH number of the susceptible human host
    IH number of the infected human host
    RH number of the recovered human host
    K sum of the total human host
    SV density of the susceptible mosquito vectors
    IV density of the infected mosquito vectors
    N sum of the total mosquito vectors density
    ¯β1 biting rate of an infected vector on the susceptible human
    ¯β2 infection incidence between infected and susceptible hosts
    β3 infection ratio between infected hosts and susceptible vectors
    η1 determines the level at which the force of infection saturates
    η2 determines the level at which the force of infection saturates
    η3 determines the level at which the force of infection saturates
    γ the conversion rate of infected hosts to recovered hosts
    μ natural death rate of human
    Λ birth or immigration of human
    m natural death rate of mosquito vectors
    d disease-induced mortality of infected hosts

     | Show Table
    DownLoad: CSV

    Obviously, we can get that there always exists a compact positively invariant set for model (1.1) as follows

    Γ0={(SH,IH,RH,SV,IV)R5+:SH+IH+RHK,SV+IVΛm}. (1.2)

    The incidence rate has various forms and plays an important role in the study of epidemic dynamics. In addition to the saturated incidence used in model (1.1) of this paper, other forms of the incidence rate have been widely used. For example, Chong, Tchuenche, and Smith [11] studied a mathematical model of avian influenza with half-saturated incidence rate SbIbHb+Ib, SHIaHa+Ia and ShImHm+Im. In addition, Li et al. [12] also carried out numerical analysis of friteral order pine wilt disease model with bilinear incidence rate ShIv and IhSv. In order to make the model (1.1) have wider research significance and apply to more infectious diseases, we consider replacing the saturated incidence rate with the general incidence rate ϕ1(IV), ϕ2(IH) and ϕ3(IH), and then give the epidemic model with the general incidence as follows

    {˙SH=μ(KSH)¯β1ϕ1(IV)SH¯β2ϕ2(IH)SH+dIH,˙IH=¯β1ϕ1(IV)SH+¯β2ϕ2(IH)SHωIH,˙RH=γIHμRH,˙SV=Λβ3Φ3(IH)SVmSV˙IV=β3ϕ3(IH)SVmIV. (1.3)

    Furthermore, the incidence rate in model (1.3) are assumed to meet the following conditions

    (A1) ϕ1(0)=ϕ2(0)=ϕ3(0)=0,

    (A2) ϕ1(IV)0,ϕ2(IH)0,ϕ3(IH)0, IV,IH0,

    (A3) 0(IVϕ1(IV))m0,0(IHϕ2(IH))m0,0(IHϕ3(IH))m0, where m0 is a positive constant.

    By looking at the model (1.3), the following equation is valid, dNdt=ΛmN, that indicates, N(t)Λm as t. Note that SH+IH+RH=K, SV+IV=Λm, this means that RH=KSHIH, SV=ΛmIV, let ω=d+μ+γ, thus the host population and pathogen population system are equivalent to the following system

    {˙SH=μ(KSH)¯β1ϕ1(IV)SH¯β2ϕ2(IH)SH+dIH,˙IH=¯β1ϕ1(IV)SH+¯β2ϕ2(IH)SHωIH,˙IV=β3ϕ3(IH)(ΛmIV)mIV. (1.4)

    Noise is ubiquitous in real life, and the spread of infectious diseases will inevitably be affected by the environment and other external factors. With the unpredictable environment, some key parameters in the infectious disease model are inevitably affected by external environmental factors. Therefore, in order to more accurately describe the transmission process, we use a stochastic model to describe and predict the epidemic trend of diseases. For the perturbation term of the parameter, two methods are commonly used, including linear function of Gaussian noise and mean-reverting stochastic process [13,14,15,16,17]. For Gaussian noise, when the time interval is very small, the variance of the parameter will become infinite, indicating that the parameter has changed greatly in a short time, which is unreasonable [18]. So we mainly consider the mean-reverting Ornstein–Uhlenbeck process. Let βi(i=1,2) take the following form

    dβi(t)=αi(¯βiβi(t))dt+σidBi(t), (1.5)

    where αi represent the speed of reversal and σi represent the intensity of fluctuation. Solve the Eq (1.5), we can get

    βi(t)=eαitβi(0)+¯βi(1eαit)+σit0eαi(ts)dBi(s),

    where βi(0) is the initial value of βi(t). For arbitrary initial value βi(0), βi(t) follows a Gaussian distribution βi(t)N(¯βi,σ2i2αi)(t). Furthermore, setting βi(0)=¯βi, then the average value of βi(t) satisfies

    ¯βi(t)=1tt0βi(s)ds=¯βi+1tt0σiαi(1eαi(st))dBi(s),

    it is known that the mathematical expectation and variance of βi(t) are ¯βi and σ2it3+O(t2), respectively, where O(t2) is the higher order infinitesimal of t2. Obviously, the variance becomes zero instead of infinity as t0. This shows the universality of the Ornstein–Uhlenbeck process. Moreover, in order to ensure the positivity of the parameter values after adding the perturbation, the log-normal Ornstein-Uhlenbeck process for the noise perturbation to the transmission rates β1 and β2 of the system (1.4) is used, then following stochastic model is obtained

    {dSH(t)=[μ(KSH(t))β1ϕ1(IV(t))SH(t)β2(t)ϕ2(IH(t))SH(t)+dIH(t)]dt,dIH(t)=[β1(t)ϕ1(IV(t))SH(t)+β2(t)ϕ2(IH(t))SH(t)ωIH(t)]dt,dIV(t)=[β3ϕ3(IH(t))(ΛmIV(t))mIV(t)]dt,dlogβ1(t)=α1(log¯β1logβ1(t))dt+σ1dB1(t),dlogβ2(t)=α2(log¯β2logβ2(t))dt+σ2dB2(t). (1.6)

    In this paper, we extend the saturated incidence rate of model (1.1) to the general incidence ϕ1(IV), ϕ2(IH) and ϕ3(IH) to obtain models (1.3) and (1.4), and investigate the global asymptotic stability of the equilibrium point of model (1.3). Furthermore, we choose to modify the parameter β1 and β2 to satisfy the log-normal Ornstein-Uhlenbeck process to obtain the stochastic model (1.6), and study its stationary distribution, exponential extinction, probability density function near the quasi-equilibrium point and other dynamic properties.

    The rest of this article is organized as follows. In Section 2, some necessary mathematical symbols and lemmas are introduced. In Section 3, some conclusions of deterministic model (1.3) are obtained and the global stability of equilibrium point in this model is proved. In Section 4, we obtain some theoretical results for the stochastic system (1.6) where we prove the existence of a unique global positive solution for the stochastic system (1.6). In addition, through the ergodic properties of parameters βi(t),i=1,2 and the construction of a series of suitable Lyapunov functions, sufficient criterion for the existence of stationary distribution is obtained, which indicates that the disease in the system will persist. Next, we have sufficient conditions for the disease to go extinct. Further, we solve the corresponding matrix equation to obtain an expression for the probability density function near the quasi-local equilibrium point of the stationary distribution. Next, in Section 5, some theoretical results are verified by several numerical simulations. Finally, several conclusions are given in Section 6.

    To make it easier to understand, denote Rn+={(y1,y2,...yn)Rn|yj>0,1jn}. In represents the n-dimensional unit matrix. IA denotes the indicator function of set A, and it means that when xA,IA=1, otherwise, IA=0. If A is a matrix or vector, then AT stands for its inverse matrix, and A1 stands for its inverse matrix.

    Lemma 2.1. (Itˆo's formula [19]) Consider the n-dimensional stochastic differential equation

    dx(t)=f(t)dt+g(t)dB(t), (2.1)

    where B(t)=(B1(t),B2(t),...,Bn(t)) and it represents n-dimensional Brownian motion defined on a complete probability space, let L act on a function VC2,1(Rn×R+;R), then we have

    dV(x(t),t)=LV(x(t),t)dt+Vx(x(t),t)g(t)dB(t),a.s.,

    where

    LV(x(t),t)=Vt(x(t),t)+Vx(x(t),t)f(t)+12trace(gT(t)Vxx(x(t),t)g(t)),

    it represents the differential operator, and

    Vt=Vt,Vx=(Vx1,...,Vxn),Vxx=(2Vxixj)n×n.

    Lemma 2.2. (Ma et al. [20]) Letting ϕ(λ)=λn+a1λn1+a2λn2++an1λ+an is the characteristic polynomial of the square matrix A, the matrix A is called a Hurwitz matrix if and only if all characteristic roots of A are negative real parts, that is equivalent to the following conditions being true

    Hk=|a1a3a5a2k11a2a4a2k20a1a3a2k301a2a2k4000ak|>0,

    k=1,2,,n, among them j>n, replenishing definition aj=0.

    Lemma 2.3. ([21]) For five-dimension algebraic equation G20+LΘ+ΘLT=0, where Θ is a symmetric matrix, G0=diag(1,0,0,0,0) and

    L=(l1l2l3l4l51000001000001000000l6).

    If l1>0,l2>0,l3>0,l4>0 and l1l2l3l23l21l4>0, then the symmetric matrix Θ is a positive semi-definite matrix. Thus, we have

    Θ=(l1l4l2l3l0l3l000l3l0l1l0l3l0l1l000l1l0l3l1l2ll4000000),

    where l=2(l4l21l1l2l3+l23).

    Lemma 2.4. ([22,23]) For n-dimension stochastic process (1.6), X(t)Rn and its initial value X(0)Rn, if there is a bounded closed domain U in Rn with a regular boundary and

    lim inft+1tt0P(τ,X(0),U)dτ>0,a.s.,

    in which P(τ,X(0),U) represents the transition probability of X(t), then X(t) has an invariant probability measure on Rn, then it admits at least one stationary distribution.

    In this section, we focus on the local stability of the equilibrium point of the deterministic model (1.3). Initially, we verify the existence and uniqueness of equilibria model (1.3). We can calculate the basic reproduction number of the deterministic model (1.3) by the next generation method [24], define

    F=(¯β2ϕ2(0)K¯β1ϕ1(0)K00),V=(ω0β3Λϕ3(0)mm),

    therefore, the next generation matrix is

    FV1=(¯β2Kϕ2(0)ω+¯β1β3ΛKϕ1(0)ϕ3(0)m2ω¯β1ϕ1(0)Km00),

    then, the basic reproduction number for system (1.3) is obtained

    R0=ρ(FV1)=¯β2Kϕ2(0)ω+¯β1β3ΛKϕ1(0)ϕ3(0)m2ω.

    Based on the key value of the basic reproduction number R0, the conditions for the existence of local equilibrium point for the model (1.3) can be found.

    Theorem 3.1. The disease-free equilibrium E0 of model (1.3) is E0=(SH0, 0, 0, SV0, 0)=(K, 0, 0, Λm, 0) which always exists. If R0>1, there is a unique local equilibrium E=(SH, IH, RH, SV, IV).

    After finding the conditions for the existence of the equilibrium point of model (1.3), next we verify that the global stability of equilibria.

    Theorem 3.2. (i) If R0<1, the disease-free equilibrium point E0 is globally asymptotically stable. If R0>1, E0 is unstable. (ii) If R0>1, the endemic equilibrium point E is globally asymptotically stable.

    Remark 3.2 The global stability of E0 in this theorem can be referred to the method in the literature [10]. By constructing a series of suitable Lyapunov functions, we prove the global stability of E.

    Initially, we verify that the stochastic model (1.6) has a unique global positive solution. This provides preparation for the dynamic behavior of the model. For model (1.6), it is easy to see that

    Γ={(SH,IH,IV,β1,β2)R5+:SH+IH<K,IV<Λm}

    is the positive invariant set, and the subsequent research will be discussed in Γ.

    Theorem 4.1. For any initial value (SH(0),IH(0),IV(0),β1(0),β2(0))Γ, there exists a unique solution (SH(t),IH(t),IV(t),β1(t),β2(t)) of system (1.6) and the solution will remain in Γ with probability one (a.s.).

    The stationary distribution of stochastic model (1.6) plays a key role in regulating the dynamics of disease and analyzing the sustainable development of disease. Next, sufficient conditions for the existence of stationary distribution will be obtained. We define

    Rs0=~β2Kϕ2(0)ω+~β1β3ΛKϕ1(0)ϕ3(0)m2ω,

    where

    ~β1=¯β1eσ2120α1,~β2=¯β2eσ2212α2.

    Theorem 4.2. If Rs0>1, then stochastic system (1.6) has a stationary distribution.

    Remark 4.2 The Theorem 4.2 is proved by constructing the Lyapunov functions. It is observed that when Rs0>1, a stationary distribution exists and the disease will be endemic for a long period of time.

    Furthermore, disease propagation and extinction are two major areas of research in stochastic system dynamics. After establishing the conditions under which a disease reaches a stable state, it is also essential to understand the conditions under which the disease becomes extinct. In addition, we discuss the sufficient condition for disease extinction in the model (1.6), define

    RE0=R0+mR0(eσ21α12eσ214α1+1)12+ϕ2(0)K¯β2(eσ22α22eσ224α2+1)12min{m,¯β2ϕ2(0)KR0}.

    Theorem 4.3. If RE0<1, then the disease of the system (1.6) will become exponentially extinct with probability 1.

    Remark 4.3 Theorem 4.3 gives the sufficient condition for the exponential extinction of diseases IH, IV. From the expressions of Rs0 and RE0, the relationships Rs0R0 and RE0R0 are deduced, and the equal sign holds if and only if σ1=σ2=0.

    Additionally, the density function of a continuous distribution is essential to understanding a stochastic system, making the precise determination of its expression a crucial challenge. Through the matrix analysis method, we have successfully derived the expression for the probability density function in the vicinity of the equilibrium point for model (1.6). Linearize the model (1.6) before calculate the probability density function. Define a quasi-endemic equilibrium point P=(SH,IH,IV,logβ1,logβ2), it satisfies

    {μKμSβ1ϕ1(IV)SHβ2ϕ2(IH)S+dIH=0,β1ϕ1(IV)SH+β2ϕ2(IH)SωIH=0,β3ϕ3(IH)(ΛmIV)mIV=0,α1(log¯β1logβ1)=0,α2(log¯β2logβ2)=0. (4.1)

    Let (z1,z2,z3,x1,x2)T=(SHSH,IHIH,IVIV,logβ1logβ1,logβ2logβ2)T, then system (4.1) can be linearized around P as follows

    {dz1=[a11z1a12z2a13z3a14x1a15x2]dt,dz2=[a21z1a22z2+a13z3+a14x1+a15x2]dt,dz3=[a32z2a33z3]dt,dx1=a44x1dt+σ1dB1(t),dx2=a55x2dt+σ2dB2(t), (4.2)

    where a11=μ+¯β1ϕ1(IV)+¯β2ϕ2(IH), a12=¯β2ϕ2(IH)SHd, a13=¯β1ϕ1(IV)SH, a14=¯β1ϕ1(IV)SH, a15=¯β2ϕ2(IH)SH, a21=¯β1ϕ1(IV)+¯β2ϕ2(IH), a22=ω¯β2ϕ2(IH)SH, a32=β3ϕ3(IH)(ΛmIV), a33=β3ϕ3(IH)+m, a44=α1, a55=α2. Apparently, aij>0.

    By denoting

    A=(a11a12a13a14a15a21a22a13a14a150a32a3300000a4400000a55), G=(000σ1σ2),
    B(t)=(0,0,0,B1(t),B2(t))T, Z(t)=(z1,z2,z3,x1,x2)T.

    Then system (4.1) can be expressed as

    dZ(t)=AZ(t)dt+GdB(t), (4.3)

    then, the solution of (4.3) can be calculated as

    X(t)=eAtX(0)+t0eA(ts)GdB(t),

    since t0eA(ts)GdB(t) obeys a normal distribution N(0,ˆΣ(t)) at time t, where ˆΣ(t)=t0eA(ts)GGTeAT(ts)ds, then, we can get X(t)N(eAtX(0),ˆΣ(t)).

    First, we need to verify that matrix A is Hurwitz matrix [25], the characteristic polynomial of A can be obtained as follows

    φA(λ)=|λ+a11a12a13a14a15a21λ+a22a13a14a150a32λ+a3300000λ+a4400000λ+a55|=(λ+a44)(λ+a55)|λ+a11a12a13a21λ+a22a230a32λ+a33|.

    Obviously there are λ1=a44,λ2=a55, other characteristic roots can also be found with negative real part according to Section 3. Therefore, matrix A is a Hurwitz matrix by Lemma 2.2. According to the stability theory of zero solution to the general linear equation [20], we have

    limt+eAtX(0)=0,
    Σlimt+ˆΣ=limt+t0eA(ts)GGTeAT(ts)ds=+0eATtG2eAtdt.

    Based on the solution of Gardiner [26], it follows

    G2+AΣ+ΣAT=0. (4.4)

    Second, we will solve the Eq (4.4) to get the exact expression of probability density function near the quasi-endemic equilibrium.

    Theorem 4.4. For any initial value (SH(0),IH(0),IV(0),β1(0),β2(0))Γ, if R0>1 and mμ+β3ϕ3(IH)>0, then the stationary distribution of stochastic system (1.6) near the P approximately admits a normal probability density function as follows

    Φ(SH,IH,IV,logβ1,logβ2)=(2π)52|Σ|12exp[12(SHSH,IHIH,IVIV,logβ1log¯β1,logβ2log¯β2)Σ1(SHSH,IHIH,IVIV,logβ1log¯β1,logβ2log¯β2)T].

    The exact expression of covariance matrix Σ is shown in the proof.

    Remark 4.4 In Theorem 4.4, by defining the quasi-endemic equilibrium P, we derive an exact expression of probability density function of the stationary distribution around a quasi-positive equilibrium P.

    In order to illustrate the above theoretical results, we perform several numerical simulations in this section. Consider bilinear incidence rate ϕ1(IV)=IV, ϕ2(IH)=IH, ϕ3(IH)=IH, letting xi(t)=logβi(t)logˉβi,i=1,2, then according to the method in [27], the following is the corresponding discretized equation for the system (1.6)

    {Sj+1H=SjH+[μ(KSjH)¯β1exj1IjVSjH+¯β2exj2IjHSjH+dIjH]Δt,Ij+iH=IjH+[¯β1exj1IjVSjH+¯β2exj2IjHSjHωIjH]Δt,Ij+1V=IjV+[β3IjH(ΛmIjV)mIjV]Δt,xj+11=xj1α1xj1Δt+σ1Δtξ1,j+σ212(ξ21,j1)Δt,xj+12=xj2α2xj2Δt+σ2Δtξ2,j+σ222(ξ22,j1)Δt,

    let ξi,j be random variables that follow a Gaussian distribution N(0,1) for i=1,2 and j=1,2,...,n. The time interval is denoted by Δt>0. The values (SjH,IjH,IjV,xj1,xj2) correspond to the j-th iteration of the discretization equation.

    First and foremost, taking into consideration the importance of parameter selection, rationality, and the visual effectiveness of theoretical results, we choose the following appropriate parameters, referring to [10,28,29], and denoted them as Number 1

    μ=0.05,K=100,¯β1=0.15,¯β2=0.1,β3=0.1,ω=0.8,d=0.5,
    γ=0.25,Λ=5,m=0.5,α1=0.8,α2=0.8,σ1=0.1,σ2=0.1,

    after calculation, we can get mμ+β3IH=2.0391>0 and the indexes of deterministic system and stochastic system can be obtained respectively, as shown below

    R0=¯β2Kω+¯β1β3Λkm2ω=50>1, Rs0=~β2Kω+~β1β3Λkm2ω=50.0365>1,

    which satisfies the condition in Theorem 4.2. More importantly, we can calculate the quasi-endemic equilibrium and the covariance matrix Σ, which have the following forms

    (SH, IH, IV, logβ1, logβ2)=(4.6565, 15.8906, 7.6066, log0.15, log0.1),
    Σ=(0.05290.03770.00380.00930.01300.03770.03520.00310.00720.01000.00380.00310.00040.00060.00080.00930.00720.00060.006200.01300.01000.000800.0062).

    Therefore, we can get that the solution (SH(t),IH(t),IV(t),β1(t),β2(t)) obeys the normal density function

    Φ(SH,IH,IV,β1,β2)N((4.6565, 15.8906, 7.6066,0.15,0.1)T,Σ).

    The marginal density functions are as follows

    ΦSH=1.7345e9.4518(SH4.6565)2, ΦIH=2.1264e14.2045(IH15.8906)2, ΦIV=19.9471e1250(IV7.6066)2.

    Using the above parameters, we can get trajectories of SH(t), IH(t) and IV(t) respectively, which are present in Figure 1. It is used to represent the variation of the solution (SH(t),IH(t),IV(t)) in the deterministic model (1.4) and the stochastic model (1.6). Frequency histograms and marginal density function curves for SH(t),IH(t) and IV(t) are also given in the right column of the Figure 1.

    Figure 1.  The left and right columns show the trajectories of the solutions (SH(t),IH(t),IV(t)) of the stochastic and deterministic systems under perturbations σ1=0.1,σ2=0.1, as well as histograms of the solutions and the marginal density functions, respectively.

    In addition, the frequency fitted density functions and the marginal density functions for SH(t),IH(t) and IV(t) are given in Figure 2, respectively, which are highly consistent. Therefore, we deduce that the solutions (SH(t),IH(t),IV(t)) have a smooth distribution and their density functions follow a normal distribution. As we can see, the disease eventually spreads, which is consistent with Theorem 4.2.

    Figure 2.  The frequency fitting density functions and marginal density functions of SH(t),IH(t) and IV(t), respectively.

    On the other hand, we select that a part of parameters are shown below, and the remaining parameters are consistent with Number 1, ¯β1=0.015,¯β2=0.001,β3=0.015. These are denoted as Number 2. The crucial value RE0 takes the form

    RE0=R0+(eσ21α12eσ214α1+1)12+K¯β2m(eσ22α22eσ224α2+1)12=0.7986<1

    which satisfies the condition in Theorem 4.3. Figure 3 represents the trajectory of the solution (SH(t),IH(t),IV(t)), and it is clearly visible that the eventual trend of the disease is towards extinction.

    Figure 3.  The trajectory of solution (SH(t),IH(t),IV(t)) under the condition RE0<1.

    Further, by choosing the parameters in Numbers 1 and 2, respectively, the left panel in Figure 4 satisfies the condition Rs0>1 and the right panel satisfies RE0<1. It can be seen that the disease exhibits a trend towards stabilization and extinction as conditions Theorems 4.2 and 4.3 are satisfied, respectively.

    Figure 4.  The left panel shows the trajectories of the solution (SH(t),IH(t),IV(t)) in the stochastic model (1.6) for Rs0>1, and the right panel shows the trajectories of the solutions of the stochastic model (1.6) for RE0<1.

    Now, we study the effect of perturbations for a mosquito-borne epidemic model. Assuming that all parameters take the values in Number 1, we choose different reversion speed α and volatility intensity σ to plot the graphs, respectively. Taking α1=α2=0.8 and different volatility intensity as shown in the Figure 5, the icons are red line σi=0.05, blue line σi=0.1 and green line σi=0.15, i=1,2, the trends of the solution (SH(t),IH(t),IV(t)) of the stochastic model (1.6) are represented by the figure. It shows that the fluctuation decrease as the volatility intensity decreases. Then, we set the volatility intensity σ1=σ2=0.1, the reversion speed αi=0.1 as shown by the red line in the figure, the blue line shows αi=1.0, and similarly, the green line indicates αi=1.5, i=1,2, then the same insightful changes in Figure 6 indicate that the fluctuation decreases with the increase of the reversion speed.

    Figure 5.  Trajectory plots of the solution (SH(t),IH(t),IV(t)) of the stochastic model (1.6) at the reversion speed αi=0.8, i=1,2 and different volatility intensity is shown in the icon with the red line σ1=σ2=0.05, the blue line σ1=σ2=0.1 and the green line σ1=σ2=0.15.
    Figure 6.  Trajectory plots of the solution (SH(t),IH(t),IV(t)) of the stochastic model (1.6) at the volatility intensity σi=0.1, i=1,2 and different reversion is shown in the icon with the red line α1=α2=0.1, the blue line α1=α2=1 and the green line α1=α2=1.5.

    Further, we make the rest of the parameter assumptions consistent with Numbers 1 and 2, respectively, except for the the volatility intensity and reversion speed. Figures 7 and 8 depict the trends of R0,Rs0, and RE0 under different volatility intensity and different reversion speed, respectively, and the range of the two variables we choose is [0,1]. Combining the information in the two figures, it can be concluded that higher reversion speed and lower volatility intensity can make RE0 and Rs0 smaller.

    Figure 7.  Trend plots of R0,Rs0, and RE0 at fixed reversion speed α1=α2=0.8 and volatility intensity σ1[0.01,1], σ2=0.1. The rest of the parameter values in the left figure are consistent with those in Number 1, and the rest of the parameter values in the right figure are consistent with those in Number 2.
    Figure 8.  Trend plots of R0,Rs0, and RE0 at fixed volatility intensity σ1=σ2=0.1 and reversion speed α1[0.01,1], α2=0.8. The rest of the parameter values in the left figure are consistent with those in Number 1, and the rest of the parameter values in the right figure are consistent with those in Number 2.

    Next, we continue to discuss the effects of reversion speed α and volatility intensity σ on Rs0 and RE0 and their magnitude relationships under different conditions.

    (i) Assuming that α1, σ1 are the variables, and the other parameters are consistent with Number 1, The Figure 9 shows the three-dimensional chromatograms of Rs0 and RE0, which are consistent with the results of Figures 7 and 8, where Rs0 increases with increasing σ1, and decreases with increasing α1. This indicates that the disease stabilizes as the reversion speed decreases or volatility intensity increases. In addition, it can be seen that both Rs0 and RE0 are greater than 1 and RE0 is greater than Rs0 in the range where α1 belongs to [0.5,0.6] and σ1 belongs to [0.005,0.09];

    Figure 9.  Color plot of the trend of RE0 and Rs0 with variables (α1,σ1)[0.5,0.6]×[0.005,0.09], with the rest of the parameters consistent with those in Number 1.

    (ii) Conditionally the same as in (i), we set the other parameters are consistent with Number 2, it is noted that RE0 increases with the increase of σ1 and decreases with the increase of α1 in Figure 10, implying that the diseases in the stochastic model (1.6) tend to become extinct when the volatility intensity decreases. Moreover, in the parameter range of the plot, both Rs0 and RE0 are less than 1, and RE0 is greater than Rs0.

    Figure 10.  Color plot of the trend of RE0 and Rs0 with variables (α1,σ1)[0.5,0.6]×[0.005,0.09], with the rest of the parameters consistent with those in Number 2.

    Next, we will discuss the mean first passage time, the moment a stochastic process first transitions from one state to another is termed the first passage time (FPT) [30]. The mean first passage time (MFPT) is then defined as the average of these first passage times [31]. Starting with an initial value of (SH(0),IH(0),IV(0)), we aim to examine the time it takes for the system to evolve from this initial state to either a stationary state (MFPT1) or to an extinction state (MFPT2).

    Then we define τ1 as the FPT from the initial state to the persistent state, and τ2 as the FPT from the initial state to the extinct state

    τ1=inf{t:SH<SH,IH>IH,IV>IV},
    τ2=inf{t:IH0<0.0001,IV0<0.0001}.

    Then we have

    MFPT1=E(τ1), MFPT2=E(τ2).

    Using Monte Carlo numerical simulation method, if SH(nΔt)<SH, IH(nΔt)>IH, IV(nΔt)>IV, then τ1=nΔt, assuming that the number of simulations is N, then

    MFPT1=Ni=1niΔtN.

    Similarly, if IH0<0.0001 and IV0<0.0001, then τ2=mΔt and

    MFPT2=Ni=1niΔtN.

    Here, we set N = 2000, σi and αi, i=1,2 are random variables. Figures 11 and 12 depict the relationship between MFPT1 and MFPT2 and the speed of reversion αi and the volatility intensity σi, i=1,2 in stochastic system (1.6) with the bilinear incidence rate, respectively. Figure 11 reveals the values of MFPT1 with N=2000,σi[0.01,0.1] and αi=4,5,6, i=1,2, respectively. It shows that MFPT1 decreases with decreasing reversion speed αi or increasing voltility intensity σi, implying that the disease is much easier to arrive the stable state. Similarly, Figure 12 shows the trend of MFPT2 at N=2000, σi[0.01,0.1] and αi=0.1,0.15,0.2. Through the figure it can be noted that MFPT2 increases with αi decrease and σi increase.

    Figure 11.  The mean first passage time for transitioning from the initial state values (SH(0),IH(0),IV(0)) = (3, 1, 1) to the state of stationary with σi[0.01,0.1], αi=4,5,6, i=1,2. The other fixed parameter values are consistent with those in Number 1.
    Figure 12.  The mean first passage time for transitioning from the initial state values (SH(0),IH(0),IV(0)) = (5,200, 10) to the state of extinction with σi[0.01,0.1], αi=0.1,0.15,0.2, i=1,2. The other fixed parameter values are consistent with those in Number 2.

    We mainly develop a stochastic model, coupled with the general incidence rate and Ornstein-Uhlenbeck process, to study the dynamic of infectious disease spread, which includes the stationary distribution and probability density function. In view of our analysis, we can draw the following conclusions

    (i) For the deterministic epidemic model, two equilibria and the basic reproduction number R0 are obtained, and their global asymptotic stability are deduced. Specificaly, the endemic equilibrium is globally asymptotically stable if R0>1, and the disease free equilibrium is globally asymptotically stable if R0<1.

    (ii) Considering that the spread of infectious disease is inevitably affected by environmental perturbations, we propose a stochastic model with general incidence and the Ornstein-Uhlenbeck process. By constructing a series of appropriate Lyapunov functions, the stationary distribution of model (1.6) is derived and we establish the sufficient criterion for the existence of the extinction. Specifically, the innovation of this paper is that we obtain a precise expression of the distribution around its quasi-positive equilibrium P by solving a difficult five-dimensional matrix equation, which is quite challenging.

    (iii) We also verify some conclusions of this paper by several numerical simulations.

    When Rs0>1, i.e., the parameters satisfy the condition of Theorem 4.2, we obtain trajectory plots of the solutions of the deterministic model (1.4) and the stochastic model (1.6), as well as the corresponding frequency histograms and edge density functions, and as shown in Figures 1 and 2, the disease eventually persists. This provides some verification of Theorem 4.2 of the theoretical results.

    Also, from Figure 3, we can further find that when RE0<1, the population strengths decrease with time and eventually converge to zero, which implies extinction of the disease. Figure 4 also further illustrates these points. The theoretical result of Theorem 4.3 is visualized through Figures 3 and 4.

    In addition, we also investigate the effect of perturbations and give Figures 5 and 6 to depict the effect of trends with different reversion speed and different volatility intensity. It can be seen that the fluctuation decreases as the volatility intensity decrease and the reversion speed increase.

    Then, correlation plots of Rs0, RE0 with reversion speed and volatility intensity are obtained in Figures 710. We conculde that higher reversion speed and lower volatility intensity can make RE0 and Rs0 more smaller.

    Finally, Figures 11 and 12 visually demonstrate the relationship between MFPT and the αi and σi, i=1,2 in a stochastic system (1.6) with bilinear incidence. We can see that if voltility intensity σi is much bigger (or the reversion speed αi is much smaller), then the disease is more easier to arrive the stable state. This is consistent with the results of the above conclusions.

    The authors declare they have not used Artificial Intelligence (AI) tools in the creation of this article.

    The authors would like to thank the anonymous referee for his/her useful suggestions. This work is supported by the Fundamental Research Funds for the Central Universities (No. 24CX03002A).

    The authors declare that they have no conflict of interest.

    Proof. The equilibria of system (1.3) satisfy

    {μ(KSH)¯β1ϕ1(IV)SH¯β2ϕ2(IH)SH+dIH=0,¯β1ϕ1(IV)SH+¯β2ϕ2(IH)SHωIH=0,γIHμRH=0,Λβ3Φ3(IH)SVmSV=0,β3ϕ3(IH)SVmIV=0. (A.1)

    Notice that

    RH=KSHIH, SV=ΛmIV,

    and we have

    {μ(KSH)¯β1ϕ1(IV)SH¯β2ϕ2(IH)SH+dIH=0,¯β1ϕ1(IV)SH+¯β2ϕ2(IH)SHωIH=0,β3ϕ3(IH)(ΛmIV)mIV=0. (A.2)

    Obviously E0=(SH0, 0, 0, SV0, 0)=(K, 0, 0, Λm, 0) always exists.

    On the other hand, we have

    {SH=K(1+γμ)IH,IV=β3ϕ3(IH)Λm2+mβ3ϕ3(IH)IH=¯β1ϕ1(IV)SHω+¯β2ϕ2(IH)SHω.

    Therefore, IH[0,μKμ+γ], let

    H(IH)=¯β1ϕ1(IV)SHω+¯β2ϕ2(IH)SHωIH,

    then by calculation, H(0)=0,H(μKμ+γ)=μKμ+γ<0, and

    H(IH)=¯β1ϕ1(IV)SHωm2β3ϕ3(IH)Λ(m2+mβ3ϕ3(IH))2(1+γμ)¯β1ϕ1(IV)ω+¯β2ϕ2(IH)SHω(1+γμ)¯β2ϕ2(IH)ω1.

    If R0>1, then

    H(0)=¯β2Kϕ2(0)ω+¯β1β3ΛKϕ1(0)ϕ3(0)m2ω1=R01>0,
    H(μKμ+γ)=(1+γμ)[¯β1ϕ1(IV(μKμ+γ))ω+¯β2ϕ2(μKμ+γ)ω]1<0,

    therefore, there exists a point ξ such that H(IH)>0 on [0,ξ) and H(IH)<0 on (ξ,μKμ+γ], i.e., H(IH) is monotonically increasing on [0,ξ) and monotonically decreasing on (ξ,μKμ+γ].

    Hence, there is a unique IH(0,μKμ+γ) such that H(IH)=0, which implies that when R0>1, system (1.4) has a unique endemic equilibrium E=(SH,IH,RH,SV,IV). This completes the proof.

    Proof. (i) Through calculation, Jacobian matrix of model (1.3) is obtained as follows

    J(SH,IH,RH,SVIV)=(μ¯β1ϕ1(IV)¯β2ϕ2(IH)d¯β2ϕ2(IH)SH00¯β1ϕ1(IV)SH¯β1ϕ1(IV)+¯β2ϕ2(IH)¯β2ϕ2(IH)SHω00¯β1ϕ2(IV)SH0γμ000β3Φ3(IH)SV0β3Φ3(IH)m00β3Φ3(IH)SV0β3Φ3(IH)m).

    Substituting E0 into the matrix J to get J0

    J0=(μd¯β2Kϕ2(0)00¯β1ϕ1(0)K0¯β2ϕ2(0)Kω00¯β1ϕ1(0)K0γμ000Λmβ3ϕ3(0)0m00Λmβ3ϕ3(0)00m),

    the corresponding characteristic polynomial is as follows

    ϕJ0(λ)=(λ+μ)(λ+μ)(λ+m)(λ+m)(λ+(ω¯β2Φ2(0))),

    obviously, if R0<1, then ω¯β2Φ2(0)>0, according to the Routh-Hurwitz criterion, there are only negative real part characteristic roots, so the disease-free equilibrium E0 is locally asymptotically stable.

    If R0>1, then ω¯β2Φ2(0)<0, this indicates that J0 has the eigenvalue of the positive real part, so the disease-free equilibrium E0 is unstable.

    Next we prove the global attractiveness of E0, define

    V=SHKKlogSHK+IH+SVΛmΛmlogmSVΛ+IV,

    Using Itˆo's formula for the above equation, we get

    LV=μK+dIHμSH¯β1Φ1(IV)SH¯β2Φ2(IH)SHμK2SHdIHKSH+¯β1Φ1(IV)K+¯β2Φ2(IH)K+μK+¯β1Φ1(IV)SH+¯β2Φ2(IH)SHωIH+Λβ3Φ3(IH)SVmSVΛ2mSV+β3Φ3(IH)Λm+Λ+β3Φ3(IH)SVmIV2μK+dIHμSHμK2SHdIHKSH+¯β1Φ1(0)IVK+¯β2Φ2(0)IHKωIHmSVΛ2mSV+β3Φ3(0)IHΛm+2ΛmIV,

    according to the method in [10], it is easy to see that SHK as t, SVΛm as t, and IH, IV0 as t. Then, LV0 and equal to 0 when it takes E0. Therefore, according to the LaSalle invariance principle, we conclude that when R0<1, E0 is globally asymptotically stable.

    (ii) According to the method in [32], the positive equilibrium point E of the model (1.3) satisfies the following system of equations

    {μK=μSH+(¯β1Φ1(IV)+¯β2Φ2(IH))SHdIH,(¯β1Φ1(IV)+¯β2Φ2(IH))SH=ωIH,γIH=μRH,Λ=β3Φ3(IH)SV+mSV,β3Φ3(IH)SV=mIV. (A.3)

    Define

    V1=SHSHSHlogSHSH+IHIHIHlogIHIH,

    after calculation

    d(SHlogSHSH)dt=SH(μKSH+μ+¯β1Φ1(IV)+¯β2Φ2(IH)dIHSH)=SH((μ+¯β1Φ1(IV)+¯β2Φ2(IH))SHdIHSH+μ+¯β1Φ1(IV)+¯β2Φ2(IH)dIHSH)=SH((μ+¯β1Φ1(IV)+¯β2Φ2(IH))SHSH+¯β1Φ1(IV)Φ1(IV)Φ1(IV)+¯β2Φ2(IH)Φ2(IH)Φ2(IH))+μSH(1SHSH)+SHSH(dIHdIH),
    d(IHlogIHIH)dt=¯β1Φ1(IV)SHSHΦ1(IV)IHSHΦ1(IV)IH¯β2Φ2(IH)SHSHΦ2(IH)IHSHΦ2(IH)IH+ωIH,
    d(SH+IH)dt=μKμSH+dIHωIH=(μ+¯β1Φ1(IV)+¯β2Φ2(IH))SHdIHμSH+dIH(¯β1Φ1(IV)+¯β2Φ2(IH))SHIHIH=μSH(1SHSH)+(¯β1Φ1(IV)+¯β2Φ2(IH))SH(1IHIH)d(IHIH),

    then we have

    dV1dt=μSH(1SHSH)+(¯β1Φ1(IV)+¯β2Φ2(IH))SH(1IHIH)d(IHIH)+ωIH+SH((μ+¯β1Φ1(IV)+¯β2Φ2(IH))SHSH+¯β1Φ1(IV)Φ1(IV)Φ1(IV)+¯β2Φ2(IH)Φ2(IH)Φ2(IH))+μSH(1SHSH)+SHSH(dIHdIH)¯β1Φ1(IV)SHSHΦ1(IV)IHSHΦ1(IV)IH¯β2Φ2(IH)SHSHΦ2(IH)IHSHΦ2(IH)IH=μSH(2SHSHSHSH)+¯β1Φ1(IV)SH[Φ1(IV)Φ1(IV)SHSHSHΦ1(IV)IHSHΦ1(IV)IHIHIH+3]+¯β2Φ2(IH)SH[Φ2(IH)Φ2(IH)SHSHSHΦ2(IH)IHSHΦ2(IH)IHIHIH+3]dIH(1SHSH)(1IHIH)μSH(2SHSHSHSH)+¯β1Φ1(IV)SH[Φ1(IV)Φ1(IV)logSHSHlogSHΦ1(IV)IHSHΦ1(IV)IHIHIH]+¯β2Φ2(IH)SH[Φ2(IH)Φ2(IH)logSHSHlogSHΦ2(IH)IHSHΦ2(IH)IHIHIH]=μ(SHSH)2SH+¯β1Φ1(IV)SH[Φ1(IV)Φ1(IV)logIHΦ1(IV)IHΦ1(IV)IHIH]+¯β2Φ2(IH)SH[Φ2(IH)Φ2(IH)logIHΦ2(IH)IHΦ2(IH)IHIH].

    Similarly, define

    V2=SVSVSVlogSVSV+IVIVIVlogIVIV,

    the same can be obtained

    dV2dt=mSV(1SVSV)+β3Φ3(IH)SV(1IVIV)+β3Φ3(IH)SV(SVSV+Φ3(IH)Φ3(IH))+mSV(1SVSV)β3SVΦ3(IH)IVSVΦ3(IH)IVSVΦ3(IH)+β3SVΦ3(IH)=mSV(1SVSVSVSV)+β3SVΦ3(IH)[Φ3(IH)Φ3(IH)SVSVIVSVΦ3(IH)IVSVΦ3(IH)IVIV+2]mSV(SVSV)2SV+β3SVΦ3(IH)[Φ3(IH)Φ3(IH)logIVΦ3(IH)IVΦ3(IH)IVIV].

    Next, define

    V3=V1+¯β1Φ1(IV)SH¯β3Φ3(IH)SVV2,

    we can get

    dV3dtμSH(SHSH)2SHm¯β1Φ1(IV)SH¯β3Φ3(IH)+¯β2Φ2(IH)SH[Φ2(IH)Φ2(IH)logIHΦ2(IH)IHΦ2(IH)IHIH]+¯β1Φ1(IV)SH[Φ1(IV)Φ1(IV)logIHΦ1(IV)IHΦ1(IV)IHIH+Φ3(IH)Φ3(IH)logIVΦ3(IH)IVΦ3(IH)IVIV]μSH(SHSH)2SHm¯β1Φ1(IV)SH¯β3Φ3(IH)+¯β2Φ2(IH)SH[Φ2(IH)Φ2(IH)+IHΦ2(IH)IHΦ2(IH)IHIH1]+¯β1Φ1(IV)SH[Φ1(IV)Φ1(IV)+IVΦ1(IV)IVΦ1(IV)IVIV1+IHΦ3(IH)IHΦ3(IH)+Φ3(IH)Φ3(IH)IHIH1],

    by the condition (A2) and (A3), we can know

    Φ1(IV)Φ1(IV)+IVΦ1(IV)IVΦ1(IV)IVIV1=IVΦ1(IV)Φ1(IV)[(Φ1(IV)Φ1(IV))(Φ1(IV)IVΦ1(IV)IV)]0,

    Φ2(IH) and Φ3(IH) similarly satisfy the structure of the above equation, combined with SH and SV are bounded, we finally get

    dV3dtμK(SHSH)2m2Λ¯β1Φ1(IV)SH¯β3Φ3(IH)SV(SVSV)2.

    Next, we define

    V4=(SHSH+IHIH)22, V5=(RHRH)22, V6=(SVSV+IVIV)22,

    similarly calculated

    dV4dt=μ(SHSH)2(d+γ)(IHIH)2ω(SHSH)(IHIH)μ(SHSH)2(d+γ)(IHIH)2+(d+γ)2(IHIH)2+ω22(d+γ)(SHSH)2ω22(d+γ)(SHSH)2(d+γ)2(IHIH)2,

    and

    dV5dt=γ(IHIH)(RHRH)μ(RHRH)2μ2(RHRH)2+γ22μ(IHIH)2,

    and

    dV6dt=m(SVSV)2m(IVIV)22m(SVSV)(IVIV)=m(SVSV)2m(IVIV)2+m2(IVIV)2+2m(SVSV)2m(SVSV)2m2(IVIV)2.

    Finally, define

    V=V3+A1V4+A2V5+A3V6,

    then

    dVdt(μKA1ω22(d+γ))(SHSH)2(d+γ2A1γ22μA2)(IHIH)2μ2A2(RHRH)2(m2Λ¯β1Φ1(IV)SH¯β3Φ3(IH)SVA3m)(SVSV)2m2A3(IVIV)2,

    take

    A1=μ(d+γ)Kω2, A2γ2μ=d+γ2A1, A3=m2Λ¯β1Φ1(IV)SH2m¯β3Φ3(IH)SV,

    we get

    dVdtμ2K(SHSH)2d+γ4A1(IHIH)2μ2A2(RHRH)2m2Λ¯β1Φ1(IV)SH2¯β3Φ3(IH)SV(SVSV)2m2A3(IVIV)2,

    the proof is done.

    Proof. It is obvious that the coefficients of the system is locally Lipschitz continuous, so there is a unique local solution (SH(t),IH(t),IV(t),β1(t),β2(t)) on t[0,τe), where τe represents explosion time. To show that the solution is global, according to the method in [33], we just need to verify that τe=+ a.s..

    Choose k0 be a sufficiently large integer for every component of (SH(0),IH(0),IV(0),β1(0),β2(0)) within the interval [1k0,k0]. For each integer kk0, define the stopping time as

    τk=inf{t[0,τe)|min(SH(t),IH(t),IV(t),β1(t),β2(t))1k or max(SH(t),IH(t),IV(t),β1(t),β2(t))k}.

    It can be seen that τk is monotonically increasing with respect to k. Then define inf{}=+ and τ=limt+τk. It is clearly visible that the solution is global due to the fact that τ<τe a.s., τ= leading to τe=. Next, we will prove τ= by the contradiction method. Assuming τ<+ a.s., then there are ε0(0,1) and T>0 such that P(τT)>ε, so there is a positive integer k1>k0 that makes

    P(τkT)ε, kk1.

    Define a non-negative C2-function V(SH,IH,IV,β1,β2) as follows

    V=SH1logSH+IH1logIH+IV1logIV+(KSHIH)1log(KSHIH)+(ΛmIV)1log(ΛmIV)+β11logβ1+β21logβ2.

    Applying Itˆo's formula to V, then we can get

    LV=μ(KSH)β1ϕ1(IV)SHβ2ϕ2(IH)SH+dIHμKSH+μ+β1ϕ1(IV)+β2ϕ2(IH)dIHSH+β1ϕ1(IV)SH+β2ϕ2(IH)SHωIHβ1ϕ1(IV)SHIHβ2ϕ2(IH)SHIH+ω+β3ϕ3(IH)(ΛmIV)mIVβ3ϕ3(IH)ΛmIV+β3ϕ3(IV)+m+1ΛmIV(β3ϕ3(IH)(ΛmIV)mIV)β3ϕ3(IH)(ΛmIV)+mIV+1K(SH+IH)(μ(KSH)+(dω)IH)μ(KSH)(dω)IH+β1(α1log¯β1α1logβ1+12σ21)α1(log¯β1logβ1)+β2(α2log¯β2α2logβ2+12σ22)α2(log¯β2logβ2)μK+dIH+μ+β1ϕ1(IV)+β2ϕ2(IH)+ω+Λmβ3ϕ3(IH)+m+2β3ϕ3(IH)+μ+γIH+β1(α1log¯β1α1logβ1+12σ21)α1(log¯β1logβ1)+β2(α2log¯β2α2logβ2+12σ22)α2(log¯β2logβ2).

    By the condition A3, we can know that

    ϕ1(IV)ϕ1(0)IV,ϕ2(IH)ϕ2(0)IH,ϕ3(IH)ϕ3(0)IH,

    then, we obtain

    LVμK+2μ+ω+m+β1ϕ1(0)IV+[β2ϕ2(0)+Λmβ3ϕ3(0)+2β3ϕ3(0)+γ]IH+β1(α1log¯β1α1logβ1+12σ21)α1(log¯β1logβ1)+β2(α2log¯β2α2logβ2+12σ22)α2(log¯β2logβ2)μK+2μ+ω+m+β1ϕ1(0)Λm+[β2ϕ2(0)+Λmβ3ϕ3(0)+2β3ϕ3(0)+γ]K+β1(α1log¯β1α1logβ1+12σ21)α1(log¯β1logβ1)+β2(α2log¯β2α2logβ2+12σ22)α2(log¯β2logβ2):=H(β1,β2).

    It's easy to see H(β1,β2) as β1+,β10,β2+,β20, so there's a positive constant H0 that makes LVH0. Integrating on both sides and taking the expectation, then

    0EW(SH(τkT),IH(τkT),IV(τkT),β1(τkT),β2(τkT))=EW(SH(0),IH(0),IV(0),β1(0),β2(0))+EτnT0LV(SH(τ),IH(τ),IV(τ),β1(τ),β2(τ))dτEV(SH(0),IH(0),IV(0),β1(0),β2(0))+H0T.

    One gets that for any ζGk, W(SH(τk,ζ),IH(τk,ζ),IV(τk,ζ),β1(τk,ζ),β2(τk,ζ)) will larger than (ek1k)(ek1+k), so

    EW(SH(0),IH(0),IV(0),β1(0),β2(0))+H0TEW(SH(τkT),IH(τkT),IV(τkT),β1(τkT),β2(τkT))E[IGk(ζ)W(SH(τkT),IH(τkT),IV(τkT),β1(τkT),β2(τkT))]P(Gk(ζ))W(SH(τk,ζ),IH(τk,ζ),IV(τk,ζ),β1(τk,ζ),β2(τk,ζ))ε0[(ek1k)(ek1+k)].

    Since is an arbitrary constant, it can be contradictory by making

    Therefore a.s., i.e., . Then system (1.6) has a unique global solution on .

    Proof. The theorem will be proved next in the following two steps.

    Step 1. Construct Lyapunov functions

    Using It's formula, we obtain

    Define a function , where are given in subsequent calculation. Using It's formula, then

    By the condition , notice that

    which can deduce

    (A.4)

    similarly, one gets

    (A.5)

    Combining (A.4) and (A.5), we have

    Let and satisfy the following equalities

    then

    where

    By Holder inequality, for any positive constant , the following equations are true

    If take to be

    then we can get

    where

    Next we define

    applying It's formula to , it leads to

    where

    Next, define

    then, we have

    where

    Choose is a large enough positive constant, let

    where satisfying the following inequality

    Notice that has a minimum value in the interior of because as tends to the boundary of . Ultimately, establish a non-negative -function as follows

    then we obtain

    (A.6)

    Step 2. Set up the closed set

    where is a small enough constant and the complement of can be divided into nine small sets as follows

    then the following results hold

    Case 1: , then

    Case 2: , then

    Case 3: , then

    Case 4: , then

    Case 5: , then

    Case 6: , then

    Case 7: , then

    Case 8: , then

    Case 9: , then

    According to the discussion of cases above, we can know that

    in other words, let is a positive constant that makes

    For any initial value , integrating the inequality (A.6) and taking the expectation, we get

    (A.7)

    One gets that is ergodic according to [34,35], then we can get that

    hence

    Then letting and taking infimum to (A.7) it follows

    which means

    According to the Lemma 2.4, we can conclude that when system (1.6) has a stationary distribution on .

    Proof. Define a -function by

    where . Applying It's formula to , then we have

    Integrating both sides of this equation from 0 to and dividing by , we get

    (A.8)

    According to the ergodicity of , then

    (A.9)

    Letting , and submitting (A.9) into (A.8), then inequailty (A.8) becomes

    where

    If ,

    will be true which indicates

    this means the disease will die out exponentially.

    Proof. Step 1 Consider the following equation

    (A.10)

    where .

    Let , where

    then

    Let , where

    and

    Due to , let , where

    and

    in which

    By using the methodology in [36,37], the standard transformation matrix of has the following form

    where

    Define , then we can get

    in which

    Let , we can equivalently transform the Eq (A.10) into

    (A.11)

    where , let , then (A.11) becomes

    then we obtian

    where . We can obtain that the matrix is a positive semi-definite matrix, the exact expression of is as follows

    Step 2 Consider the following equation

    (A.12)

    where .

    Let , where

    then

    Let , where

    then

    Similarly, due to , let , where

    and

    in which

    The standard transformation matrix of has the following form

    where

    Define , then we can get

    in which

    Let , the equation (A.12) can be equivalently transformed into

    (A.13)

    where , let , then (A.13) becomes

    then we obtian

    where , and we can obtain that the matrix is a positive semi-definite matrix, the exact expression of is as follows

    Finally, . Obviously, the matrix is a positive definite matrix. The proof is complete.



    Conflict of Interest



    The authors declare no conflict of interest.

    [1] Drachman DA (2005) Do we have brain to spare? Neurology 64: 2004-2005. doi: 10.1212/01.WNL.0000166914.38327.BB
    [2] Beul SF, Hilgetag CC (2019) Neuron density fundamentally relates to architecture and connectivity of the primate cerebral cortex. Neuroimage 189: 777-792. doi: 10.1016/j.neuroimage.2019.01.010
    [3] Cunningham EL, McGuinness B, Herron B, et al. (2015) Dementia. Ulster Med J 84: 79-87.
    [4] Alzheimer's Disease International (2014) Dementia in the Asia Pacific Region. Alzheimer's Disease International Report, London Available from: https://www.alzint.org/resource/dementia-in-the-asia-pacific-region/.
    [5] Europe A (2019)  Dementia in Europe yearbook 2019: Estimating the prevalence of dementia in Europe Luxembourg: Alzheimer Europe, 1-102.
    [6] Alzheimer's disease facts and figures (2020) Alzheimer's Dement. Alzheimer's Association Report 2020 Available from: https://doi.org/10.1002/alz.12068.
    [7] Kumar CTS, Shaji KS, Varghese M, et al. (2019)  Dementia in India 2020 Cochin: Alzheimer's and Related Disorders Society of India (ARDSI), 1-96.
    [8] Barnes DE, Yaffe K (2011) The projected effect of risk factor reduction on Alzheimer's disease prevalence. Lancet Neurol 10: 819-828. doi: 10.1016/S1474-4422(11)70072-2
    [9] Fleming R, Zeisel J, Bennett K (2020)  World Alzheimer Report 2020: Design Dignity Dementia: dementia-related design and the built environment Volume 1: Case Studies London, England: Alzheimer's Disease International, 1-248.
    [10] Trevisan K, Cristina-Pereira R, Silva-Amaral D, et al. (2019) Theories of Aging and the Prevalence of Alzheimer's Disease. Biomed Res Int 2019: 1-9. doi: 10.1155/2019/9171424
    [11] Alzheimer A (1987) About a peculiar disease of the cerebral cortex. By Alois Alzheimer, 1907 (Translated by L Jarvik and H Greenson). Alzheimer Dis Assoc Disord 1: 3-8.
    [12] Tiraboschi P, Hansen LA, Thal LJ, et al. (2004) The importance of neuritic plaques and tangles to the development and evolution of AD. Neurology 62: 1984-1989. doi: 10.1212/01.WNL.0000129697.01779.0A
    [13] Carter J, Lippa CF (2001) Beta-Amyloid, Neuronal Death and Alzheimers Disease. Curr Mol Med 1: 733-737. doi: 10.2174/1566524013363177
    [14] Medina M, Hernández F, Avila J (2016) New features about tau function and dysfunction. Biomolecules 6: 21. doi: 10.3390/biom6020021
    [15] Castellani RJ, Perry G (2019) Tau biology, tauopathy, traumatic brain injury, and diagnostic challenges. J Alzheimers Dis 67: 447-467. doi: 10.3233/JAD-180721
    [16] Zvěřová M (2019) Clinical aspects of Alzheimer's disease. Clin Biochem 72: 3-6. doi: 10.1016/j.clinbiochem.2019.04.015
    [17] Yang Y, Kandiah N, Kim S, et al. (2020) Clinical aspects of neurobehavioral symptoms of dementia. Dement Neurocogn Disord 19: 54-64. doi: 10.12779/dnd.2020.19.2.54
    [18] Raz L, Knoefel J, Bhaskar K (2016) The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab 36: 172-186. doi: 10.1038/jcbfm.2015.164
    [19] Livingston G, Huntley J, Sommerlad A, et al. (2020) Dementia prevention, intervention, and care: report of the Lancet Commission. Lancet 396: 413-446. doi: 10.1016/S0140-6736(20)30367-6
    [20] Kelly AM (2018) Exercise-induced modulation of neuroinflammation in models of Alzheimer's disease. Brain Plast 4: 81-94. doi: 10.3233/BPL-180074
    [21] Saha P, Sen N (2019) Tauopathy: A common mechanism for neurodegeneration and brain aging. Mech Ageing Dev 178: 72-79. doi: 10.1016/j.mad.2019.01.007
    [22] Hernandez F, Avila J (2007) Tauopathies. Cell Mol Life Sci 64: 2219-2233. doi: 10.1007/s00018-007-7220-x
    [23] Šimić G, Babić Leko M, Wray S, et al. (2016) Tau protein hyperphosphorylation and aggregation in Alzheimer's disease and other tauopathies, and possible neuroprotective strategies. Biomolecules 6: 6. doi: 10.3390/biom6010006
    [24] Vickers JC, Dickson TC, Adlard PA, et al. (2000) The cause of neuronal degeneration in Alzheimer's disease. Prog Neurobiol 60: 139-165. doi: 10.1016/S0301-0082(99)00023-4
    [25] Serrano-Pozo A, Frosch MP, Masliah E, et al. (2011) Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 1: a006189. doi: 10.1101/cshperspect.a006189
    [26] Wenk GL (2003) Neuropathologic Changes in Alzheimer's Disease. J Clin Psychiatry 64: 7-10.
    [27] Priller C, Bauer T, Mitteregger G, et al. (2006) Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci 26: 7212-7221. doi: 10.1523/JNEUROSCI.1450-06.2006
    [28] Turner PR, O'connor K, Tate WP, et al. (2003) Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog Neurobiol 70: 1-32. doi: 10.1016/S0301-0082(03)00089-3
    [29] Sastre M, Steiner H, Fuchs K, et al. (2001) Presenilin-dependent γ-secretase processing of β-amyloid precursor protein at a site corresponding to the S3 cleavage of Notch. EMBO Rep 2: 835-841. doi: 10.1093/embo-reports/kve180
    [30] Selkoe DJ (2002) Deciphering the genesis and fate of amyloid beta-protein yields novel therapies for Alzheimer disease. J Clin Invest 110: 1375-1381. doi: 10.1172/JCI0216783
    [31] Dingledine R, Borges K, Bowie D, et al. (1999) The glutamate receptor ion channels. Pharmacol Rev 51: 7-62.
    [32] Hu M, Liu QS, Chang KT, et al. (2002) Nicotinic regulation of CREB activation in hippocampal neurons by glutamatergic and nonglutamatergic pathways. Mol Cell Neurosci 21: 616-625. doi: 10.1006/mcne.2002.1202
    [33] Yakel JL (2013) Cholinergic receptors: functional role of nicotinic ACh receptors in brain circuits and disease. Pflügers Archi 465: 441-450. doi: 10.1007/s00424-012-1200-1
    [34] Hussain I, Powell D, Howlett DR, et al. (1999) Identification of a novel aspartic protease (Asp 2) as β-secretase. Mol Cell Neurosci 14: 419-427. doi: 10.1006/mcne.1999.0811
    [35] Seubert P, Oltersdorf T, Lee MG, et al. (1993) Secretion of β-amyloid precursor protein cleaved at the amino terminus of the β-amyloid peptide. Nature 361: 260-263. doi: 10.1038/361260a0
    [36] Hashimoto M, Rockenstein E, Crews L, et al. (2003) Role of protein aggregation in mitochondrial dysfunction and neurodegeneration in Alzheimer's and Parkinson's diseases. Neuromolecular Med 4: 21-36. doi: 10.1385/NMM:4:1-2:21
    [37] Peng C, Trojanowski JQ, Lee VM (2020) Protein transmission in neurodegenerative disease. Nat Rev Neurol 16: 199-212. doi: 10.1038/s41582-020-0333-7
    [38] Mattson MP, Guo ZH, Geiger JD (1999) Secreted form of amyloid precursor protein enhances basal glucose and glutamate transport and protects against oxidative impairment of glucose and glutamate transport in synaptosomes by a cyclic GMP-mediated mechanism. J Neurochem 73: 532-537. doi: 10.1046/j.1471-4159.1999.0730532.x
    [39] Snyder EM, Nong Y, Almeida CG, et al. (2005) Regulation of NMDA receptor trafficking by amyloid-β. Nat Neurosci 8: 1051-1058. doi: 10.1038/nn1503
    [40] Gu Z, Liu W, Yan Z (2009) β-Amyloid impairs AMPA receptor trafficking and function by reducing Ca2+/calmodulin-dependent protein kinase II synaptic distribution. J Biol Chem 284: 10639-10649. doi: 10.1074/jbc.M806508200
    [41] Tozaki H, Matsumoto A, Kanno T, et al. (2002) The inhibitory and facilitatory actions of amyloid-β peptides on nicotinic ACh receptors and AMPA receptors. Biochem Biophys Rese Communn 294: 42-45. doi: 10.1016/S0006-291X(02)00429-1
    [42] Liskowsky W, Schliebs R (2006) Muscarinic acetylcholine receptor inhibition in transgenic Alzheimer-like Tg2576 mice by scopolamine favours the amyloidogenic route of processing of amyloid precursor protein. Int J Dev Neurosci 24: 149-156. doi: 10.1016/j.ijdevneu.2005.11.010
    [43] Bojarski L, Herms J, Kuznicki J (2008) Calcium dysregulation in Alzheimer's disease. Neurochem Int 52: 621-633. doi: 10.1016/j.neuint.2007.10.002
    [44] Van der Kant R, Goldstein LS, Ossenkoppele R (2020) Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat Rev Neurosci 21: 21-35. doi: 10.1038/s41583-019-0240-3
    [45] Ramesh G, MacLean AG, Philipp MT (2013) Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm 2013: 480739.
    [46] Alberghina L, Colangelo AM (2006) The modular systems biology approach to investigate the control of apoptosis in Alzheimer's disease neurodegeneration. BMC Neurosci 7: S2. doi: 10.1186/1471-2202-7-S1-S2
    [47] Gunn AP, Wong BX, Johanssen T, et al. (2016) Amyloid-β peptide Aβ3pE-42 induces lipid peroxidation, membrane permeabilization, and calcium influx in neurons. J Biol Chem 291: 6134-6145. doi: 10.1074/jbc.M115.655183
    [48] Gan X, Huang S, Wu L, et al. (2014) Inhibition of ERK-DLP1 signaling and mitochondrial division alleviates mitochondrial dysfunction in Alzheimer's disease cybrid cell. Biochim Biophys Acta 1842: 220-231. doi: 10.1016/j.bbadis.2013.11.009
    [49] Hooper C, Killick R, Lovestone S (2008) The GSK3 hypothesis of Alzheimer's disease. J Neurochem 104: 1433-1439. doi: 10.1111/j.1471-4159.2007.05194.x
    [50] Town T, Zolton J, Shaffner R, et al. (2002) p35/Cdk5 pathway mediates soluble amyloid-β peptide-induced tau phosphorylation in vitro. J Neurosci Res 69: 362-372. doi: 10.1002/jnr.10299
    [51] Das A, Dikshit M, Nath C (2005) Role of molecular isoforms of acetylcholinesterase in learning and memory functions. Pharmacol Biochem Behav 81: 89-99. doi: 10.1016/j.pbb.2005.02.006
    [52] Tiwari P, Dwivedi S, Singh MP, et al. (2013) Basic and modern concepts on cholinergic receptor: A review. Asian Pac J Trop Dis 3: 413-420. doi: 10.1016/S2222-1808(13)60094-8
    [53] Leonard BE (2004) Pharmacotherapy in the treatment of Alzheimer's disease: an update. World Psychiatry 3: 84-88.
    [54] Gutteridge JM, Halliwell B (2000) Free radicals and antioxidants in the year 2000: a historical look to the future. Ann N Y Acad Sci 899: 136-147. doi: 10.1111/j.1749-6632.2000.tb06182.x
    [55] Uttara B, Singh AV, Zamboni P, et al. (2009) Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol 7: 65-74. doi: 10.2174/157015909787602823
    [56] Mancuso C, Scapagini G, Curro D, et al. (2007) Mitochondrial dysfunction, free radical generation and cellular stress response in neurodegenerative disorders. Front Biosci 12: 1107-1123. doi: 10.2741/2130
    [57] Beal MF (1998) Mitochondrial dysfunction in neurodegenerative diseases. Biochim Biophys Acta 1366: 211-223. doi: 10.1016/S0005-2728(98)00114-5
    [58] Fernie AR, Carrari F, Sweetlove LJ (2004) Respiratory metabolism: glycolysis, the TCA cycle and mitochondrial electron transport. Curr Opin Plant Biol 7: 254-261. doi: 10.1016/j.pbi.2004.03.007
    [59] Turrens JF (2003) Mitochondrial formation of reactive oxygen species. J Physiol 552: 335-344. doi: 10.1113/jphysiol.2003.049478
    [60] Halliwell B (1992) Reactive oxygen species and the central nervous system. J Neurochem 59: 1609-1623. doi: 10.1111/j.1471-4159.1992.tb10990.x
    [61] Reddy PH, Beal MF (2008) Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer's disease. Trends Mol Med 14: 45-53. doi: 10.1016/j.molmed.2007.12.002
    [62] Reddy PH, Tripathi R, Troung Q, et al. (2012) Abnormal mitochondrial dynamics and synaptic degeneration as early events in Alzheimer's disease: implications to mitochondria-targeted antioxidant therapeutics. Biochim Biophys Acta 1822: 639-649. doi: 10.1016/j.bbadis.2011.10.011
    [63] Reddy PH, McWeeney S, Park BS, et al. (2004) Gene expression profiles of transcripts in amyloid precursor protein transgenic mice: up-regulation of mitochondrial metabolism and apoptotic genes is an early cellular change in Alzheimer's disease. Hum Mol Genet 13: 1225-1240. doi: 10.1093/hmg/ddh140
    [64] Tran M, Reddy PH (2021) Defective Autophagy and Mitophagy in Aging and Alzheimer's Disease. Front Neurosci 14: 612757. doi: 10.3389/fnins.2020.612757
    [65] Pradeepkiran JA, Reddy PH (2020) Defective mitophagy in Alzheimer's disease. Ageing Res Rev 64: 101191. doi: 10.1016/j.arr.2020.101191
    [66] Reddy PH (2011) Abnormal tau, mitochondrial dysfunction, impaired axonal transport of mitochondria, and synaptic deprivation in Alzheimer's disease. Brain Res 1415: 136-148. doi: 10.1016/j.brainres.2011.07.052
    [67] Garbarino VR, Orr ME, Rodriguez KA, et al. (2015) Mechanisms of oxidative stress resistance in the brain: lessons learned from hypoxia tolerant extremophilic vertebrates. Arch Biochem Biophys 576: 8-16. doi: 10.1016/j.abb.2015.01.029
    [68] Gerlach M, Ben-Shachar D, Riederer P, et al. (1994) Altered brain metabolism of iron as a cause of neurodegenerative diseases? J Neurochem 63: 793-807. doi: 10.1046/j.1471-4159.1994.63030793.x
    [69] Chance B, Sies H, Boveris A (1979) Hydroperoxide metabolism in mammalian organs. Physiol Rev 59: 527-605. doi: 10.1152/physrev.1979.59.3.527
    [70] Hirai K, Aliev G, Nunomura A, et al. (2001) Mitochondrial abnormalities in Alzheimer's disease. J Neurosci 21: 3017-3023. doi: 10.1523/JNEUROSCI.21-09-03017.2001
    [71] Tuppo EE, Arias HR (2005) The role of inflammation in Alzheimer's disease. Int J Biochem Cell Biol 37: 289-305. doi: 10.1016/j.biocel.2004.07.009
    [72] Chao CC, Hu S, Sheng WS, et al. (1996) Cytokine-stimulated astrocytes damage human neurons via a nitric oxide mechanism. Glia 16: 276-284. doi: 10.1002/(SICI)1098-1136(199603)16:3<276::AID-GLIA10>3.0.CO;2-X
    [73] Biber K, Vinet J, Boddeke HW (2008) Neuron-microglia signaling: chemokines as versatile messengers. J Neuroimmunol 198: 69-74. doi: 10.1016/j.jneuroim.2008.04.012
    [74] Perkins ND (2007) Integrating cell-signalling pathways with NF-κB and IKK function. Nat Rev Mol Cell Biol 8: 49-62. doi: 10.1038/nrm2083
    [75] O'Neill LA, Kaltschmidt C (1997) NF-kB: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci 20: 252-258. doi: 10.1016/S0166-2236(96)01035-1
    [76] Friedlander RM (2003) Apoptosis and caspases in neurodegenerative diseases. N Engl J Med 348: 1365-1375. doi: 10.1056/NEJMra022366
    [77] Katsimpardi L, Lledo PM (2018) Regulation of neurogenesis in the adult and aging brain. Curr Opin Neurobiol 53: 131-138. doi: 10.1016/j.conb.2018.07.006
    [78] Anjum I, Fayyaz M, Wajid A, et al. (2018) Does obesity increase the risk of dementia: a literature review. Cureus 10: e2660.
    [79] Verdile G, Keane KN, Cruzat VF, et al. (2015) Inflammation and oxidative stress: the molecular connectivity between insulin resistance, obesity, and Alzheimer's disease. Mediators Inflamm 2015: 105828. doi: 10.1155/2015/105828
    [80] Nameni G, Farhangi MA, Hajiluian G, et al. (2017) Insulin deficiency: a possible link between obesity and cognitive function. Int J Dev Neurosci 59: 15-20. doi: 10.1016/j.ijdevneu.2017.02.008
    [81] Steen E, Terry BM, Rivera EJ, et al. (2005) Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease—is this type 3 diabetes? J Alzheimers Dis 7: 63-80. doi: 10.3233/JAD-2005-7107
    [82] Craft S (2005) Insulin resistance syndrome and Alzheimer's disease: age-and obesity-related effects on memory, amyloid, and inflammation. Neurobiol Aging 26: 65-69. doi: 10.1016/j.neurobiolaging.2005.08.021
    [83] Dye L, Boyle NB, Champ C, et al. (2017) The relationship between obesity and cognitive health and decline. Proc Nutr Soc 76: 443-454. doi: 10.1017/S0029665117002014
    [84] Skoog I, Gustafson D (2003) Hypertension, hypertension-clustering factors and Alzheimer's disease. Neurol Res 25: 675-680. doi: 10.1179/016164103101201986
    [85] Luchsinger JA, Reitz C, Patel B, et al. (2007) Relation of diabetes to mild cognitive impairment. Arch Neurol 64: 570-575. doi: 10.1001/archneur.64.4.570
    [86] Luchsinger JA, Tang MX, Shea S, et al. (2004) Hyperinsulinemia and risk of Alzheimer disease. Neurology 63: 1187-1192. doi: 10.1212/01.WNL.0000140292.04932.87
    [87] Tiehuis AM, Van der Graaf Y, Visseren FL, et al. (2008) Diabetes increases atrophy and vascular lesions on brain MRI in patients with symptomatic arterial disease. Stroke 39: 1600-1603. doi: 10.1161/STROKEAHA.107.506089
    [88] Li ZG, Zhang W, Sima AA (2007) Alzheimer-like changes in rat models of spontaneous diabetes. Diabetes 56: 1817-1824. doi: 10.2337/db07-0171
    [89] Ho L, Qin W, Pompl PN, et al. (2004) Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer's disease. FASEB J 18: 902-904. doi: 10.1096/fj.03-0978fje
    [90] Bennett RG, Hamel FG, Duckworth WC (2003) An insulin-degrading enzyme inhibitor decreases amylin degradation, increases amylin-induced cytotoxicity, and increases amyloid formation in insulinoma cell cultures. Diabetes 52: 2315-2320. doi: 10.2337/diabetes.52.9.2315
    [91] Shiiki T, Ohtsuki S, Kurihara A, et al. (2004) Brain insulin impairs amyloid-β (1–40) clearance from the brain. J Neurosci 24: 9632-9637. doi: 10.1523/JNEUROSCI.2236-04.2004
    [92] Lee HJ, Seo HI, Cha HY, et al. (2018) Diabetes and Alzheimer's disease: mechanisms and nutritional aspects. Clin Nutr Res 7: 229-240. doi: 10.7762/cnr.2018.7.4.229
    [93] Xi YD, Li XY, Ding J, et al. (2013) Soy isoflavone alleviates Aβ1-42-induced impairment of learning and memory ability through the regulation of RAGE/LRP-1 in neuronal and vascular tissue. Curr Neurovasc Res 10: 144-156. doi: 10.2174/1567202611310020007
    [94] Deane R, Wu Z, Sagare A, et al. (2004) LRP/amyloid β-peptide interaction mediates differential brain efflux of Aβ isoforms. Neuron 43: 333-344. doi: 10.1016/j.neuron.2004.07.017
    [95] Lloret A, Monllor P, Esteve D, et al. (2019) Obesity as a risk factor for Alzheimer's disease: implication of leptin and glutamate. Front Neurosci 13: 508. doi: 10.3389/fnins.2019.00508
    [96] Bird TD (2008) Genetic aspects of Alzheimer disease. Genet Med 10: 231-239. doi: 10.1097/GIM.0b013e31816b64dc
    [97] Kang J, Lemaire HG, Unterbeck A, et al. (1987) The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor. Nature 325: 733-736. doi: 10.1038/325733a0
    [98] Weidemann A, König G, Bunke D, et al. (1989) Identification, biogenesis, and localization of precursors of Alzheimer's disease A4 amyloid protein. Cell 57: 115-126. doi: 10.1016/0092-8674(89)90177-3
    [99] Scheuner D, Eckman C, Jensen M, et al. (1996) Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nat Med 2: 864-870. doi: 10.1038/nm0896-864
    [100] Sherrington R, Rogaev EI, Liang Y, et al. (1995) Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature 375: 754-760. doi: 10.1038/375754a0
    [101] Theuns J, Del-Favero J, Dermaut B, et al. (2000) Genetic variability in the regulatory region of presenilin 1 associated with risk for Alzheimer's disease and variable expression. Hum Mol Genet 9: 325-331. doi: 10.1093/hmg/9.3.325
    [102] Citron M, Westaway D, Xia W, et al. (1997) Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. Nat Med 3: 67-72. doi: 10.1038/nm0197-67
    [103] Moehlmann T, Winkler E, Xia X, et al. (2002) Presenilin-1 mutations of leucine 166 equally affect the generation of the Notch and APP intracellular domains independent of their effect on Abeta 42 production. Proc Natl Acad Sci USA 99: 8025-8030. doi: 10.1073/pnas.112686799
    [104] Levy-Lahad E, Wasco W, Poorkaj P, et al. (1995) Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science 269: 973-977. doi: 10.1126/science.7638622
    [105] Sherrington R, Froelich S, Sorbi S, et al. (1996) Alzheimer's disease associated with mutations in presenilin 2 is rare and variably penetrant. Hum Mol Genet 5: 985-988. doi: 10.1093/hmg/5.7.985
    [106] Kovacs DM, Fausett HJ, Page KJ, et al. (1996) Alzheimer-associated presenilins 1 and 2: neuronal expression in brain and localization to intracellular membranes in mammalian cells. Nat Med 2: 224-229. doi: 10.1038/nm0296-224
    [107] Corder EH, Saunders AM, Strittmatter WJ, et al. (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 261: 921-923. doi: 10.1126/science.8346443
    [108] Nicoll JA, Roberts GW, Graham DI (1995) Apolipoprotein E epsilon 4 allele is associated with deposition of amyloid beta-protein following head injury. Nat Med 1: 135-137. doi: 10.1038/nm0295-135
    [109] Liu Y, Laakso MP, Karonen JO, et al. (2002) Apolipoprotein E polymorphism and acute ischemic stroke: a diffusion- and perfusion-weighted magnetic resonance imaging study. J Cereb Blood Flow Metab 22: 1336-1342. doi: 10.1097/01.WCB.0000033200.58646.B3
    [110] Horsburgh K, McCulloch J, Nilsen M, et al. (2000) Increased neuronal damage and apoE immunoreactivity in human apolipoprotein E, E4 isoform-specific, transgenic mice after global cerebral ischaemia. Eur J Neurosci 12: 4309-4317.
    [111] Huang Y (2006) Molecular and cellular mechanisms of apolipoprotein E4 neurotoxicity and potential therapeutic strategies. Curr Opin Drug Discov Devel 9: 627-641.
    [112] Mahley RW, Weisgraber KH, Huang Y (2006) Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including Alzheimer's disease. Proc Natl Acad Sci USA 103: 5644-5651. doi: 10.1073/pnas.0600549103
    [113] Nagy Z, Esiri MM, Jobst KA, et al. (1995) Influence of the apolipoprotein E genotype on amyloid deposition and neurofibrillary tangle formation in Alzheimer's disease. Neurosci 69: 757-761. doi: 10.1016/0306-4522(95)00331-C
    [114] Gibson GE, Haroutunian V, Zhang H, et al. (2000) Mitochondrial damage in Alzheimer's disease varies with apolipoprotein E genotype. Ann Neurol 48: 297-303. doi: 10.1002/1531-8249(200009)48:3<297::AID-ANA3>3.0.CO;2-Z
    [115] Yamanaka A, Sakurai T, Katsumoto T, et al. (1999) Chronic intracerebroventricular administration of orexin—A to rats increases food intake in daytime, but has no effect on body weight. Brain Res 849: 248-252. doi: 10.1016/S0006-8993(99)01905-8
    [116] Chow M, Cao M (2016) The hypocretin/orexin system in sleep disorders: preclinical insights and clinical progress. Nat Sci Sleep 8: 81-86. doi: 10.2147/NSS.S114969
    [117] Tsuneki H, Wada T, Sasaoka T (2012) Role of orexin in the central regulation of glucose and energy homeostasis. Endocr J 59: 365-374. doi: 10.1507/endocrj.EJ12-0030
    [118] Ben J, Soares FM, Cechetti F, et al. (2009) Exercise effects on activities of Na+, K+-ATPase, acetylcholinesterase and adenine nucleotides hydrolysis in ovariectomized rats. Brain Res 1302: 248-255. doi: 10.1016/j.brainres.2009.09.013
    [119] Feng P, Vurbic D, Wu Z, et al. (2008) Changes in brain orexin levels in a rat model of depression induced by neonatal administration of clomipramine. J Psychopharmacol 22: 784-791. doi: 10.1177/0269881106082899
    [120] Liguori C, Romigi A, Nuccetelli M, et al. (2014) Orexinergic system dysregulation, sleep impairment, and cognitive decline in Alzheimer disease. JAMA Neurol 71: 1498-1505. doi: 10.1001/jamaneurol.2014.2510
    [121] Osorio RS, Ducca EL, Wohlleber ME, et al. (2016) Orexin—A is associated with increases in cerebrospinal fluid phosphorylated-tau in cognitively normal elderly subjects. Sleep 39: 1253-1260. doi: 10.5665/sleep.5846
    [122] Slats D, AHR Claassen J, Jan Lammers G, et al. (2012) Association between hypocretin-1 and amyloid-β42 cerebrospinal fluid levels in Alzheimer's disease and healthy controls. Curr Alzheimer Res 9: 1119-1125. doi: 10.2174/156720512804142840
    [123] Davies J, Chen J, Pink R, et al. (2015) Orexin receptors exert a neuroprotective effect in Alzheimer's disease (AD) via heterodimerization with GPR103. Sci Rep 5: 1-2.
    [124] Jaussent I, Bouyer J, Ancelin ML, et al. (2012) Excessive sleepiness is predictive of cognitive decline in the elderly. Sleep 35: 1201-1207. doi: 10.5665/sleep.2070
    [125] Suh SW, Han JW, Lee JR, et al. (2018) Sleep and cognitive decline: a prospective nondemented elderly cohort study. Ann Neurol 83: 472-482. doi: 10.1002/ana.25166
    [126] American Academy of Sleep Medicine (2014)  International Classification of Sleep Disorders Illinois: Darien, 1-383.
    [127] Sadeghmousavi S, Eskian M, Rahmani F, et al. (2020) The effect of insomnia on development of Alzheimer's disease. J Neuroinflammation 17: 289. doi: 10.1186/s12974-020-01960-9
    [128] Gehrman PR, Martin JL, Shochat T, et al. (2003) Sleep-disordered breathing and agitation in institutionalized adults with Alzheimer disease. Am J Geriatr Psychiatry 11: 426-433. doi: 10.1097/00019442-200307000-00005
    [129] Andrade AG, Bubu OM, Varga AW, et al. (2018) The Relationship between Obstructive Sleep Apnea and Alzheimer's Disease. J Alzheimers Dis 64: S255-S270. doi: 10.3233/JAD-179936
    [130] Pan W, Kastin AJ (2014) Can sleep apnea cause Alzheimer's disease? Neurosci Biobehav Rev 47: 656-669. doi: 10.1016/j.neubiorev.2014.10.019
    [131] Weldemichael DA, Grossberg GT (2010) Circadian rhythm disturbances in patients with Alzheimer's disease: a review. Int J Alzheimers Dis 2010: 716453.
    [132] Bhidayasiri R, Sringean J, Rattanachaisit W, et al. (2017) The sleeping brain in Parkinson's disease: A focus on REM sleep behaviour disorder and related parasomnias for practicing neurologists. J Neurol Sci 374: 32-37. doi: 10.1016/j.jns.2017.01.018
    [133] Panel CC, Watson NF, Badr MS, Belenky G, et al. (2015) Joint consensus statement of the American Academy of Sleep Medicine and Sleep Research Society on the recommended amount of sleep for a healthy adult: methodology and discussion. Sleep 38: 1161-1183. doi: 10.5665/sleep.4886
    [134] Matricciani L, Bin YS, Lallukka T, et al. (2017) Past, present, and future: trends in sleep duration and implications for public health. Sleep Health 3: 317-323. doi: 10.1016/j.sleh.2017.07.006
    [135] Lo JC, Groeger JA, Cheng GH, et al. (2016) Self-reported sleep duration and cognitive performance in older adults: a systematic review and meta-analysis. Sleep Med 17: 87-98. doi: 10.1016/j.sleep.2015.08.021
    [136] Peter-Derex L, Yammine P, Bastuji H, et al. (2015) Sleep and Alzheimer's disease. Sleep Med Rev 19: 29-38. doi: 10.1016/j.smrv.2014.03.007
    [137] Camargos EF, Louzada FM, Nóbrega OT (2013) Wrist actigraphy for measuring sleep in intervention studies with Alzheimer's disease patients: application, usefulness, and challenges. Sleep Med Rev 17: 475-488. doi: 10.1016/j.smrv.2013.01.006
    [138] Brzecka A, Leszek J, Ashraf GM, et al. (2018) Sleep disorders associated with Alzheimer's disease: a perspective. Front Neurosci 12: 330. doi: 10.3389/fnins.2018.00330
    [139] Tsolaki A, Kazis D, Kompatsiaris I, et al. (2014) Electroencephalogram and Alzheimer's disease: clinical and research approaches. Int J Alzheimers Dis 2014: 349249.
    [140] Wu YH, Feenstra MG, Zhou JN, et al. (2003) Molecular changes underlying reduced pineal melatonin levels in Alzheimer disease: alterations in preclinical and clinical stages. J Clin Endocrinol Metab 88: 5898-5906. doi: 10.1210/jc.2003-030833
    [141] Fossati S, Cam J, Meyerson J, et al. (2010) Differential activation of mitochondrial apoptotic pathways by vasculotropic amyloid-β variants in cells composing the cerebral vessel walls. FASEB J 24: 229-241. doi: 10.1096/fj.09-139584
    [142] Washington PM, Morffy N, Parsadanian M, et al. (2014) Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-beta in an Alzheimer's disease mouse model. J Neurotrauma 31: 125-134. doi: 10.1089/neu.2013.3017
    [143] Washington PM, Villapol S, Burns MP (2016) Polypathology and dementia after brain trauma: does brain injury trigger distinct neurodegenerative diseases, or should they be classified together as traumatic encephalopathy? Exp Neurol 275: 381-388. doi: 10.1016/j.expneurol.2015.06.015
    [144] Ramos-Cejudo J, Wisniewski T, Marmar C, et al. (2018) Traumatic brain injury and Alzheimer's disease: the cerebrovascular link. EBioMedicine 28: 21-30. doi: 10.1016/j.ebiom.2018.01.021
    [145] Elahi FM, Miller BL (2017) A clinicopathological approach to the diagnosis of dementia. Nat Rev Neurol 13: 457-476. doi: 10.1038/nrneurol.2017.96
    [146] Lim X, Yeo JM, Green A, et al. (2013) The diagnostic utility of cerebrospinal fluid alpha-synuclein analysis in dementia with Lewy bodies: a systematic review and meta-analysis. Parkinsonism Relat d 19: 851-858. doi: 10.1016/j.parkreldis.2013.06.008
    [147] Watson R, Colloby SJ, Blamire AM, et al. (2015) Assessment of regional gray matter loss in dementia with Lewy bodies: a surface-based MRI analysis. Am J Geriatr Psychiatry 23: 38-46. doi: 10.1016/j.jagp.2014.07.005
    [148] Chow N, Aarsland D, Honarpisheh H, et al. (2012) Comparing hippocampal atrophy in Alzheimer's dementia and dementia with Lewy bodies. Dement Geriatr Cogn Disord 34: 44-50. doi: 10.1159/000339727
    [149] Lebedev AV, Westman E, Beyer MK, et al. (2013) Multivariate classification of patients with Alzheimer's and dementia with Lewy bodies using high-dimensional cortical thickness measurements: an MRI surface-based morphometric study. J Neurol 260: 1104-1115. doi: 10.1007/s00415-012-6768-z
    [150] Watson R, O'Brien JT, Barber R, et al. (2012) Patterns of gray matter atrophy in dementia with Lewy bodies: a voxel-based morphometry study. Int Psychogeriatr 24: 532-540. doi: 10.1017/S1041610211002171
    [151] Whitwell JL, Weigand SD, Shiung MM, et al. (2007) Focal atrophy in dementia with Lewy bodies on MRI: a distinct pattern from Alzheimer's disease. Brain 130: 708-719. doi: 10.1093/brain/awl388
    [152] Watson R, Blamire AM, Colloby SJ, et al. (2012) Characterizing dementia with Lewy bodies by means of diffusion tensor imaging. Neurology 79: 906-914. doi: 10.1212/WNL.0b013e318266fc51
    [153] Galvin JE, Price JL, Yan Z, et al. (2011) Resting bold fMRI differentiates dementia with Lewy bodies vs Alzheimer disease. Neurology 76: 1797-1803. doi: 10.1212/WNL.0b013e31821ccc83
    [154] Johnson KA, Fox NC, Sperling RA, et al. (2012) Brain imaging in Alzheimer disease. Cold Spring Harb Perspect Med 2: a006213. doi: 10.1101/cshperspect.a006213
    [155] McKeith I, O'Brien J, Walker Z, et al. (2007) Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy bodies: a phase III, multicentre study. Lancet Neurol 6: 305-313. doi: 10.1016/S1474-4422(07)70057-1
    [156] Brigo F, Turri G, Tinazzi M (2015) 123I-FP-CIT SPECT in the differential diagnosis between dementia with Lewy bodies and other dementias. J Neurol Sci 359: 161-171. doi: 10.1016/j.jns.2015.11.004
    [157] Shimizu S, Hanyu H, Kanetaka H, et al. (2005) Differentiation of dementia with Lewy bodies from Alzheimer's disease using brain SPECT. Dement Geriatr Cogn Disord 20: 25-30. doi: 10.1159/000085070
    [158] Hanyu H, Shimizu S, Hirao K, et al. (2006) Comparative value of brain perfusion SPECT and [(123) I] MIBG myocardial scintigraphy in distinguishing between dementia with Lewy bodies and Alzheimer's disease. Eur J Nucl Med Mol Imaging 33: 248-253. doi: 10.1007/s00259-005-1921-x
    [159] Yoshita M, Arai H, Arai H, et al. (2015) Diagnostic accuracy of 123 I-meta-iodobenzylguanidine myocardial scintigraphy in dementia with Lewy bodies: a multicenter study. PloS One 10: e0120540. doi: 10.1371/journal.pone.0120540
    [160] Edison P, Rowe CC, Rinne JO, et al. (2008) Amyloid load in Parkinson's disease dementia and Lewy body dementia measured with [11C] PIB positron emission tomography. J Neurol Neurosurg Psychiatry 79: 1331-1338. doi: 10.1136/jnnp.2007.127878
    [161] Kantarci K, Lowe VJ, Boeve BF, et al. (2017) AV-1451 tau and β-amyloid positron emission tomography imaging in dementia with Lewy bodies. Ann Neurol 81: 58-67. doi: 10.1002/ana.24825
    [162] Coleman KK, Coleman BL, MacKinley JD, et al. (2016) Detection and differentiation of frontotemporal dementia and related disorders from alzheimer disease using the Montreal Cognitive Assessment. Alzheimer Dis Assoc Disord 30: 258-263. doi: 10.1097/WAD.0000000000000119
    [163] Kuhlmann J, Andreasson U, Pannee J, et al. (2017) CSF Aβ1–42—an excellent but complicated Alzheimer's biomarker—a route to standardisation. Clinica Chimica Acta 467: 27-33. doi: 10.1016/j.cca.2016.05.014
    [164] Lashley T, Schott JM, Weston P, et al. (2018) Molecular biomarkers of Alzheimer's disease: progress and prospects. Dis Models Mech 11: dmm031781. doi: 10.1242/dmm.031781
    [165] Morbelli S, Bauckneht M (2018) Amyloid PET imaging: standardization and integration with other Alzheimer's disease biomarkers. In Biomarkers for Alzheimer's Disease Drug Development New York, NY: Humana Press, 203-212. doi: 10.1007/978-1-4939-7704-8_13
    [166] Olsson B, Lautner R, Andreasson U, et al. (2016) CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis. Lancet Neurol 15: 673-684. doi: 10.1016/S1474-4422(16)00070-3
    [167] Guzman-Martinez L, Maccioni RB, Farías GA, et al. (2019) Biomarkers for Alzheimer's Disease. Curr Alzheimer Res 16: 518-528. doi: 10.2174/1567205016666190517121140
    [168] Gill SS, Rochon PA, Herrmann N, et al. (2005) Atypical antipsychotic drugs and risk of ischaemic stroke: population based retrospective cohort study. BMJ 330: 445. doi: 10.1136/bmj.38330.470486.8F
    [169] Giacobini E, Pepeu G (2006) Cholinesterases in human brain: the effect of cholinesterase inhibitors on Alzheimer's disease and related disorders. Brain Cholinergic Syst Health Dis 235-264. doi: 10.1201/b14486-19
    [170] Li WM, Pi RB, Chan HH, et al. (2005) Novel dimeric acetylcholinesterase inhibitor bis7-tacrine, but not donepezil, prevents glutamate-induced neuronal apoptosis by blocking N-methyl-D-aspartate receptors. J Biol Chem 280: 18179-18188. doi: 10.1074/jbc.M411085200
    [171] Hyde C, Peters J, Bond M, et al. (2013) Evolution of the evidence on the effectiveness and cost-effectiveness of acetylcholinesterase inhibitors and memantine for Alzheimer's disease: systematic review and economic model. Age Ageing 42: 14-20. doi: 10.1093/ageing/afs165
    [172] Lipton SA (2005) The molecular basis of memantine action in Alzheimer's disease and other neurologic disorders: low-affinity, uncompetitive antagonism. Curr Alzheimer Res 2: 155-165. doi: 10.2174/1567205053585846
    [173] Hynd MR, Scott HL, Dodd PR (2004) Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer's disease. Neurochem Int 45: 583-595. doi: 10.1016/j.neuint.2004.03.007
    [174] Liperoti R, Pedone C, Corsonello A (2008) Antipsychotics for the treatment of behavioral and psychological symptoms of dementia (BPSD). Curr Neuropharmacol 6: 117-124. doi: 10.2174/157015908784533860
    [175] Reynolds GP, Kirk SL (2010) Metabolic side effects of antipsychotic drug treatment—pharmacological mechanisms. Pharmacol Ther 125: 169-179. doi: 10.1016/j.pharmthera.2009.10.010
    [176] Vigen CL, Mack WJ, Keefe RS, et al. (2011) Cognitive effects of atypical antipsychotic medications in patients with Alzheimer's disease: outcomes from CATIE-AD. Am J Psychiatry 168: 831-839. doi: 10.1176/appi.ajp.2011.08121844
    [177] Ralph SJ, Espinet AJ (2018) Increased all-cause mortality by antipsychotic drugs: updated review and meta-analysis in dementia and general mental health care. J Alzheimers Dis Rep 2: 1-26. doi: 10.3233/ADR-170042
    [178] Doody RS, Raman R, Farlow M, et al. (2013) A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med 369: 341-350. doi: 10.1056/NEJMoa1210951
    [179] Coric V, Salloway S, van Dyck CH, et al. (2015) Targeting prodromal Alzheimer disease with avagacestat: a randomized clinical trial. JAMA neurology 72: 1324-1333. doi: 10.1001/jamaneurol.2015.0607
    [180] Muntimadugu E, Dhommati R, Jain A, et al. (2016) Intranasal delivery of nanoparticle encapsulated tarenflurbil: A potential brain targeting strategy for Alzheimer's disease. Eur J Pharm Sci 92: 224-234. doi: 10.1016/j.ejps.2016.05.012
    [181] Das B, Yan R (2019) A Close Look at BACE1 Inhibitors for Alzheimer's Disease Treatment. CNS Drugs 33: 251-263. doi: 10.1007/s40263-019-00613-7
    [182] Egan MF, Kost J, Voss T, et al. (2019) Randomized trial of verubecestat for prodromal Alzheimer's disease. N Engl J Med 380: 1408-1420. doi: 10.1056/NEJMoa1812840
    [183] Burki T (2018) Alzheimer's disease research: the future of BACE inhibitors. Lancet 391: 2486. doi: 10.1016/S0140-6736(18)31425-9
    [184] Henley D, Raghavan N, Sperling R, et al. (2019) Preliminary results of a trial of atabecestat in preclinical Alzheimer's disease. N Engl J Med 380: 1483-1485. doi: 10.1056/NEJMc1813435
    [185] Wong KH, Riaz MK, Xie Y, et al. (2019) Review of current strategies for delivering Alzheimer's disease drugs across the blood-brain barrier. Int J Mol Sci 20: 381. doi: 10.3390/ijms20020381
    [186] Di Benedetto G, Burgaletto C, Carta AR, et al. (2019) Beneficial effects of curtailing immune susceptibility in an Alzheimer's disease model. J Neuroinflammation 16: 166. doi: 10.1186/s12974-019-1554-9
    [187] Quitterer U, AbdAlla SN (2020) Improvements of symptoms of Alzheimers disease by inhibition of the angiotensin system. Pharmacol Res 154: 104230. doi: 10.1016/j.phrs.2019.04.014
    [188] Blurton-Jones M, Kitazawa M, Martinez-Coria H, et al. (2009) Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci USA 106: 13594-13599. doi: 10.1073/pnas.0901402106
    [189] Blurton-Jones M, Spencer B, Michael S, et al. (2014) Neural stem cells genetically-modified to express neprilysin reduce pathology in Alzheimer transgenic models. Stem Cell Res Ther 5: 46. doi: 10.1186/scrt440
    [190] Ager RR, Davis JL, Agazaryan A, et al. (2015) Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer's disease and neuronal loss. Hippocampus 25: 813-826. doi: 10.1002/hipo.22405
    [191] Martini AC, Forner S, Trujillo-Estrada L, et al. (2018) Past to future: what animal models have taught us about Alzheimer's disease. J Alzheimers Dis 64: S365-S378. doi: 10.3233/JAD-179917
    [192] Briggs R, Kennelly SP, O'Neill D (2016) Drug treatments in Alzheimer's disease. Clin Med 16: 247-253. doi: 10.7861/clinmedicine.16-3-247
    [193] Cummings J, Lee G, Ritter A, et al. (2020) Alzheimer's disease drug development pipeline: 2020. Alzheimers Dement TRCI 6: e12050.
    [194] Bennett T, Bray D, Neville MW (2014) Suvorexant, a dual orexin receptor antagonist for the management of insomnia. Pharm Ther 39: 264-266.
    [195] Equihua-Benítez AC, Guzmán-Vásquez K, Drucker-Colín R (2017) Understanding sleep-wake mechanisms and drug discovery. Expert Opin Drug Discov 12: 643-657. doi: 10.1080/17460441.2017.1329818
    [196] Steiner MA, Winrow CJ (2014) Insomnia and beyond-Exploring the therapeutic potential of orexin receptor antagonists. Frontiers Research Topics E-book series Switzerland: Frontiers Media SA, 1-219.
    [197] Pradeepkiran JA, Reddy PH (2019) Structure based design and molecular docking studies for phosphorylated tau inhibitors in Alzheimer's disease. Cells 8: 260. doi: 10.3390/cells8030260
    [198] Rêgo ML, Cabral DA, Costa EC, et al. (2019) Physical exercise for individuals with hypertension: It is time to emphasize its benefits on the brain and cognition. Clin Med Insights Cardiol 13: 1179546819839411.
    [199] Archer T, Josefsson T, Lindwall M (2014) Effects of physical exercise on depressive symptoms and biomarkers in depression. CNS Neurol Disord Drug Targets 13: 1640-1653. doi: 10.2174/1871527313666141130203245
    [200] Vina J, Sanchis-Gomar F, Martinez-Bello V, et al. (2012) Exercise acts as a drug; the pharmacological benefits of exercise. Br J Pharmacol 167: 1-12. doi: 10.1111/j.1476-5381.2012.01970.x
    [201] Daré LR, Garcia A, Neves BH, et al. (2020) One Physical Exercise Session Promotes Recognition Learning In Rats With Cognitive Deficits Related To Amyloid Beta Neurotoxicity. Brain Res 1744: 146918. doi: 10.1016/j.brainres.2020.146918
    [202] Carro E, Trejo JL, Busiguina S, et al. (2001) Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J Neurosci 21: 5678-5684. doi: 10.1523/JNEUROSCI.21-15-05678.2001
    [203] He XF, Liu DX, Zhang Q, et al. (2017) Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci 10: 144. doi: 10.3389/fnmol.2017.00144
    [204] Baker LD, Frank LL, Foster-Schubert K, et al. (2010) Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch Neurol 67: 71-79.
    [205] Lange-Asschenfeldt C, Kojda G (2008) Alzheimer's disease, cerebrovascular dysfunction and the benefits of exercise: from vessels to neurons. Exp Gerontol 43: 499-504. doi: 10.1016/j.exger.2008.04.002
    [206] Ziemann E, Zembroñ-Lacny A, Kasperska A, et al. (2013) Exercise training-induced changes in inflammatory mediators and heat shock proteins in young tennis players. J Sports Sci Med 12: 282-289.
    [207] Bobinski F, Teixeira JM, Sluka KA, et al. (2018) IL-4 mediates the analgesia produced by low-intensity exercise in mice with neuropathic pain. Pain 159: 437-450. doi: 10.1097/j.pain.0000000000001109
    [208] Liu Y, Chu JM, Yan T, et al. (2020) Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3xTg Alzheimer's disease mice. J Neuroinflammation 17: 4. doi: 10.1186/s12974-019-1653-7
    [209] Saucedo Marquez CM, Vanaudenaerde B, Troosters T, et al. (2015) High-intensity interval training evokes larger serum BDNF levels compared with intense continuous exercise. J Appl Physiol 119: 1363-1373. doi: 10.1152/japplphysiol.00126.2015
    [210] Pajonk FG, Wobrock T, Gruber O, et al. (2010) Hippocampal plasticity in response to exercise in schizophrenia. Arch Gen Psychiat 67: 133-143. doi: 10.1001/archgenpsychiatry.2009.193
    [211] Adlard PA, Perreau VM, Pop V, et al. (2005) Voluntary exercise decreases amyloid load in a transgenic model of Alzheimer's disease. J Neurosci 25: 4217-4221. doi: 10.1523/JNEUROSCI.0496-05.2005
    [212] Um HS, Kang EB, Koo JH, et al. (2011) Treadmill exercise represses neuronal cell death in an aged transgenic mouse model of Alzheimer's disease. Neurosci Res 69: 161-173. doi: 10.1016/j.neures.2010.10.004
    [213] Leem YH, Lim HJ, Shim SB, et al. (2009) Repression of tau hyperphosphorylation by chronic endurance exercise in aged transgenic mouse model of tauopathies. J Neurosci Res 87: 2561-2570. doi: 10.1002/jnr.22075
    [214] Belarbi K, Burnouf S, Fernandez-Gomez FJ, et al. (2011) Beneficial effects of exercise in a transgenic mouse model of Alzheimer's disease-like Tau pathology. Neurobiol Dis 43: 486-494. doi: 10.1016/j.nbd.2011.04.022
    [215] Alkadhi KA, Dao AT (2018) Exercise decreases BACE and APP levels in the hippocampus of a rat model of Alzheimer's disease. Mol Cell Neurosci 86: 25-29. doi: 10.1016/j.mcn.2017.11.008
    [216] Hashiguchi D, Campos HC, Wuo-Silva R, et al. (2020) Resistance Exercise Decreases Amyloid Load and Modulates Inflammatory Responses in the APP/PS1 Mouse Model for Alzheimer's Disease. J Alzheimers Dis 73: 1525-1539. doi: 10.3233/JAD-190729
    [217] Mankhong S, Kim S, Moon S, et al. (2020) Effects of Aerobic Exercise on Tau and Related Proteins in Rats with the Middle Cerebral Artery Occlusion. Int J Mol Sci 21: 5842. doi: 10.3390/ijms21165842
    [218] Khazaeinia T, Ramsey AA, Tam YK (2020) The effects of exercise on the pharmacokinetics of drugs. J Pharm Pharm Sci 3: 292-302.
    [219] Taylor D (2014) Physical activity is medicine for older adults. Postgrad Med J 90: 26-32. doi: 10.1136/postgradmedj-2012-131366
    [220] World Health Organization WHO guidelines on physical activity and sedentary behaviour: at a glance (2020) .Available from: World Health Organization. https://apps.who.int/iris/handle/10665/337001. Licence: CC BY-NC-SA 3.0 IGO, 1–17.
    [221] Spirduso WW, Francis K, MacRae P (1995)  Physical dimensions of aging USA Champaign Illinois: Human kinetics, 1-432.
    [222] Van Praag H, Shubert T, Zhao C, et al. (2005) Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci 25: 8680-8685. doi: 10.1523/JNEUROSCI.1731-05.2005
    [223] Lledo PM, Alonso M, Grubb MS (2006) Adult neurogenesis and functional plasticity in neuronal circuits. Nat Rev Neurosci 7: 179-193. doi: 10.1038/nrn1867
    [224] Erickson KI, Voss MW, Prakash RS, et al. (2011) Exercise training increases size of hippocampus and improves memory. PNAS 108: 3017-3022. doi: 10.1073/pnas.1015950108
    [225] Erickson KI, Kramer AF (2009) Aerobic exercise effects on cognitive and neural plasticity in older adults. Br J Sports Med 43: 22-24. doi: 10.1136/bjsm.2008.052498
    [226] Voss MW, Prakash RS, Erickson KI, et al. (2010) Plasticity of brain networks in a randomized intervention trial of exercise training in older adults. Front Aging Neurosci 2: 32.
    [227] Langlois F, Vu TT, Chassé K, et al. (2013) Benefits of physical exercise training on cognition and quality of life in frail older adults. J Gerontol:Series B 68: 400-404. doi: 10.1093/geronb/gbs069
    [228] Geda YE, Roberts RO, Knopman DS, et al. (2010) Physical exercise, aging, and mild cognitive impairment: a population-based study. Arch Neurol 67: 80-86.
    [229] Nuzum H, Stickel A, Corona M, et al. (2020) Potential benefits of physical activity in MCI and dementia. Behav Neurol 2020: 7807856. doi: 10.1155/2020/7807856
    [230] Burns JM, Cronk BB, Anderson HS, et al. (2008) Cardiorespiratory fitness and brain atrophy in early Alzheimer disease. Neurology 71: 210-216. doi: 10.1212/01.wnl.0000317094.86209.cb
    [231] Chen WW, Zhang X, Huang WJ (2016) Role of physical exercise in Alzheimer's disease. Biomed Rep 4: 403-407. doi: 10.3892/br.2016.607
    [232] Teri L, Gibbons LE, McCurry SM, et al. (2003) Exercise plus behavioral management in patients with Alzheimer disease: a randomized controlled trial. JAMA 290: 2015-2022. doi: 10.1001/jama.290.15.2015
    [233] Teri L, McCurry SM, Buchner DM, et al. (1998) Exercise and activity level in Alzheimer's disease: a potential treatment focus. J Rehabil Res Dev 35: 411-419.
    [234] Palleschi L, Vetta F, De Gennaro E, et al. (1996) Effect of aerobic training on the cognitive performance of elderly patients with senile dementia of Alzheimer type. Arch Gerontol Geriatr 22: 47-50. doi: 10.1016/0167-4943(96)86912-3
    [235] George EK, Reddy PH (2019) Can healthy diets, regular exercise, and better lifestyle delay the progression of dementia in elderly individuals? J Alzheimers Dis 72: S37-S58. doi: 10.3233/JAD-190232
    [236] Fitzgerald PB, Daskalakis ZJ (2013) Repetitive transcranial magnetic stimulation treatment for depressive disorders: a practical guide. Springer Science & Business Media, Germany: Springer-Verlag Berlin Heidelberg, 1-121.
    [237] Weiler M, Stieger KC, Long JM, et al. (2020) Transcranial Magnetic Stimulation in Alzheimer's disease: are we ready? eNeuro 7. doi: 10.1523/ENEURO.0235-19.2019
    [238] Abraham G, Milev R, Lazowski L, et al. (2007) Repetitive transcranial magnetic stimulation for treatment of elderly patients with depression—an open label trial. Neuropsychiatr Dis Treat 3: 919-924.
    [239] Cotelli M, Manenti R, Cappa SF, et al. (2008) Transcranial magnetic stimulation improves naming in Alzheimer disease patients at different stages of cognitive decline. Eur J Neurol 15: 1286-1292. doi: 10.1111/j.1468-1331.2008.02202.x
    [240] Cotelli M, Calabria M, Manenti R, et al. (2011) Improved language performance in Alzheimer disease following brain stimulation. J Neurol Neurosurg Psychiatry 82: 794-797. doi: 10.1136/jnnp.2009.197848
    [241] Rutherford G, Lithgow B, Moussavi Z (2015) Short and long-term effects of rTMS treatment on Alzheimer's disease at different stages: a pilot study. J Exp Neurosci 9: 43-51. doi: 10.4137/JEN.S24004
    [242] Koch G, Bonnì S, Pellicciari MC, et al. (2018) Transcranial magnetic stimulation of the precuneus enhances memory and neural activity in prodromal Alzheimer's disease. Neuroimage 169: 302-311. doi: 10.1016/j.neuroimage.2017.12.048
    [243] Rabey JM, Dobronevsky E (2016) Repetitive transcranial magnetic stimulation (rTMS) combined with cognitive training is a safe and effective modality for the treatment of Alzheimer's disease: clinical experience. J Neural Transm 123: 1449-1455. doi: 10.1007/s00702-016-1606-6
    [244] Yang YW, Pan WX, Xie Q (2020) Combined effect of repetitive transcranial magnetic stimulation and physical exercise on cortical plasticity. Neural Regen Res 15: 1986-1994. doi: 10.4103/1673-5374.282239
    [245] Ross RE, VanDerwerker CJ, Newton JH, et al. (2018) Simultaneous aerobic exercise and rTMS: Feasibility of combining therapeutic modalities to treat depression. Brain Stimul 11: 245-246. doi: 10.1016/j.brs.2017.10.019
    [246] Reed T, Kadosh RC (2018) Transcranial electrical stimulation (tES) mechanisms and its effects on cortical excitability and connectivity. J Inherit Metab Dis 41: 1123-1130. doi: 10.1007/s10545-018-0181-4
    [247] Zarifkar AH, Zarifkar A, Nami M, et al. (2018) The Comparison of the Effects of Different Transcranial Electrical Stimulation (tES) Paradigms on Beta-Amyloid (Aβ 25–35)-Induced Memory Impairment upon Morris Water Maze Task in Male Rats. J Neurol Neurosci 9: 265. doi: 10.21767/2171-6625.1000265
    [248] Steinberg F, Pixa NH, Fregni F (2019) A review of acute aerobic exercise and transcranial direct current stimulation effects on cognitive functions and their potential synergies. Front Hum Neurosci 12: 534. doi: 10.3389/fnhum.2018.00534
  • Reader Comments
  • © 2021 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(9293) PDF downloads(593) Cited by(19)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog