Skip to main content

Therapeutic non-invasive brain treatments in Alzheimer’s disease: recent advances and challenges

Abstract

Alzheimer’s disease (AD) is one of the major neurodegenerative diseases and the most common form of dementia. Characterized by the loss of learning, memory, problem-solving, language, and other thinking abilities, AD exerts a detrimental effect on both patients’ and families’ quality of life. Although there have been significant advances in understanding the mechanism underlying the pathogenesis and progression of AD, there is no cure for AD. The failure of numerous molecular targeted pharmacologic clinical trials leads to an emerging research shift toward non-invasive therapies, especially multiple targeted non-invasive treatments. In this paper, we reviewed the advances of the most widely studied non-invasive therapies, including photobiomodulation (PBM), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and exercise therapy. Firstly, we reviewed the pathological changes of AD and the challenges for AD studies. We then introduced these non-invasive therapies and discussed the factors that may affect the effects of these therapies. Additionally, we review the effects of these therapies and the possible mechanisms underlying these effects. Finally, we summarized the challenges of the non-invasive treatments in future AD studies and clinical applications. We concluded that it would be critical to understand the exact underlying mechanisms and find the optimal treatment parameters to improve the translational value of these non-invasive therapies. Moreover, the combined use of non-invasive treatments is also a promising research direction for future studies and sheds light on the future treatment or prevention of AD.

Introduction

First described in 1906 by Dr. Alois Alzheimer, Alzheimer’s disease (AD) has been recognized for more than 100 years [1]. As the most common form of dementia, AD causes progressive memory impairment and is characterized by amyloid plaques and neurofibrillary tangles [2]. There are currently estimated to be over 55 million people living with Alzheimer’s disease or other dementias all over the world, and more than 6.2 million Americans are living with Alzheimer’s disease [3]. Although scientists have made tremendous progress in better understanding the molecular mechanisms underlying amyloid-β and tau pathology in the past decades, the precise mechanisms of AD remain under hot debate, and there are no effective pharmacologic disease-modifying treatments for AD [4].

The failure of molecular targeted pharmacologic therapies has triggered increasing studies to shift toward non-invasive therapies [5]. Non-invasive brain treatment describes treatments for brain diseases that do not require an incision into the brain or tissue removal [6]. In the past decades, non-invasive therapies, including PBM therapy (also known as low-level laser therapy, light therapy), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and exercise therapy, have received increased attention as potential treatments for many brain disorders [7,8,9,10,11]. Although significant advances have been achieved in investigating the effectiveness and underlying mechanisms of these non-invasive therapies, the current understating of the exact underlying mechanism in AD is limited. This review summarizes the pathological changes of Alzheimer’s disease, provides an extensive review of the most widely studied non-invasive approaches, and discusses the primary challenges of these approaches in AD clinical applications.

Pathological changes in Alzheimer’s disease

Amyloid plaques

Amyloid plaque is one of the hallmarks of AD [12]. It is formed by the accumulation of extracellular Aβ, a 40-42 amino acid peptide derived from the amyloid precursor protein (APP) [13]. The amyloid hypothesis has been the mainstream explanation for AD’s etiology and pathogenesis since 1991 [12, 14]. As shown in Fig. 1, APP, a transmembrane protein, is cleaved by three enzymes: α, β, and γ-secretase [15]. In the normal physiological state, most of the APP (90% or more) are cleaved by α-secretase and γ-secretase, which generates sAPPα and C terminal fragments (p3, CTF 83, and AICD50). The remaining APP is cleaved by β- and γ-secretase and generates Aβ, which is rapidly removed or degraded [12]. However, under pathological conditions, most of the APP undergoes the amyloidogenic APP processing pathway, wherein Aβ generation is significantly increased and induces the formation of Aβ amyloid fibrils [16]. The accumulation of Aβ induces neurotoxicity, which finally leads to neuronal death and neurodegeneration [16].

Fig. 1
figure 1

Diagram of amyloid plaque formation. APP, a transmembrane protein, is cleaved by three enzymes, including α, β, and γ-secretase. In the normal physiological state, most of the APP (90% or more) are cleaved by α-secretase and γ-secretase, which generates sAPPα and C terminal fragments (p3, CTF 83, and AICD50). However, under pathological conditions, most of the APP undergoes the amyloidogenic APP processing pathway, wherein Aβ generation is significantly increased and induces the formation of Aβ amyloid fibrils. APP, amyloid precursor protein; sAPPα, soluble amyloid precursor protein α; sAPPβ, soluble amyloid precursor protein β; ACID, APP intracellular domain; CTF, carboxy-terminal fragment

Neurofibrillary tangles

Neurofibrillary tangles (NFTs) are another hallmark involving the pathogenesis of AD [17]. NFTs are made of abnormally hyperphosphorylated tau, a microtubule-binding protein that maintains the microtubule structure [18]. Microtubules are α and β tubulin subunits-formed hollow cylinder structures [19]. Under normal conditions, tau binds to the microtubules facilitating microtubule assembly and promoting microtubule stabilization [20]. As a significant component of the cytoskeleton, microtubules play a central role in axonal transport, cell motility, and the maintenance of cell shape [19]. In addition, the tau-associated regulation of microtubules is involved in the dynamic control of protein kinases and phosphatases [21]. In AD, the abnormally phosphorylated tau dissociates from the microtubules, and the phosphorylated tau proteins form neurofibrillary tangles (Fig. 2), which results in the interruption of axonal transports and cell signal communication [18].

Fig. 2
figure 2

Diagram of the formation of neurofibrillary tangles. Under normal conditions, tau binds to the microtubules facilitating microtubule assembly and promoting microtubule stabilization. In AD, the abnormally phosphorylated tau dissociates from the microtubules, and the phosphorylated tau proteins form neurofibrillary tangles

Mitochondrial dysfunction

The mitochondria are essential organelles in eukaryotic cells that carry out multiple essential functions, including the generation of adenosine triphosphate (ATP), intracellular signaling, biosynthesis of neuronal iron-sulfur centers and heme, regulation of cell survival, and apoptosis under various stresses [22,23,24]. To date, a large body of research has shown that mitochondrial dysfunction and impaired energy metabolism resulting from deleterious fragmentation is an early and causal event in AD [22]. Aberrant mitochondrial fragmentation, resulting from the imbalance of the fusion-fission process, plays a central role in the induction of mitochondrial dysfunction and contributes to the pathogenesis and pathology of AD [22, 23].

In the living cell, mitochondrial morphologies are far from static [25] and undergo specific mitochondrial dynamics involving coordinated cycles of mitochondrial fission and fusion [26]. Mitochondrial fission is essential for generating new mitochondria and mitochondrial quality control (removing damaged mitochondria and mitochondrial apoptosis during high cellular stress levels) [25]. As shown in Fig. 3, mitochondrial fission is mediated by Drp1, Mff, and Fis1 [27]. Drp 1 is located in the cytosol, and Fis1 and Mff are located on the outer mitochondrial membrane [27]. Mitochondrial fission occurs when the Fis1 and MFF recruit cytosolic Drp1 to the outer mitochondrial membrane [25]. In contrast, mitochondrial fusion is a process that joins two mitochondria together, which is vital in protecting mitochondrial integrity [25]. In addition, mitochondria can compensate for other mitochondria’s defects during mitochondrial fusion by sharing components to maintain their integrity [25]. Mitochondrial fusion is mediated by the Mfn1 and Mfn2 from the mitochondrial outer membranes and Opa1 in the mitochondrial inner membranes [22, 25].

Fig. 3
figure 3

Mitochondrial fission and fusion. Under healthy conditions, the healthy mitochondrial network is maintained by the balance of mitochondria fission (right) and fusion. Drp1 is located in the cytosol, and Fis1 and Mff are located on the outer mitochondrial membrane. Mitochondrial fission occurs when the Fis1 and MFF recruit cytosolic Drp1 to the outer mitochondrial membrane. Mitochondrial fusion is mediated by Mfn1 and Mfn2 from the mitochondrial outer membranes and Opa1 in the mitochondrial inner membranes

Mitochondrial fission and fusion work together to preserve mitochondrial health and function [25, 28]. Mitochondrial fission segregates the most severely damaged mitochondria and removes them through quality control, while mitochondrial fusion corrects low levels of damage by sharing components [25, 29]. In the AD brain, the balance between mitochondrial fission and fusion is disrupted by significantly increased fission protein expression and decreased fusion protein expression, which induces excessive mitochondrial fragmentation in vulnerable neurons (Fig. 3) [7, 30, 31].

In addition to abnormal mitochondrial dynamics, the role of mitochondrial bioenergetic deficits in AD has been well established [32, 33]. The high energy demand of neuronal cells suggests that mitochondrial bioenergetic deficits contribute to neuronal death in AD [34]. There are five multiprotein enzyme complexes in the mitochondrial inner membrane involved in ATP production: complex I (NADH-ubiquinone oxidoreductase), complex II (succinate-ubiquinone oxidoreductase), complex III (cytochrome bc1 complex), complex IV (cytochrome c oxidase), and complex V (ATP synthase) (Fig. 4) [22]. These five enzyme complexes’ reduced expression and activity have been found in the brain of AD patients and animals, suggesting mitochondrial bioenergetic deficit precedes Alzheimer’s pathology [32, 35, 36].

Fig. 4
figure 4

Generation of reactive oxygen species (ROS) by the mitochondria. Under physiological conditions, a small number of electrons (dotted red line) leak out of the ETC and are transferred to oxygen to produce reactive oxygen species (ROS). However, in pathological situations, mitochondrial dysfunction and the impaired mitochondrial complex activity induce significantly increased ROS production. This leads to deleterious effects and a vicious cycle causing neuroinflammation, mitochondrial damage, energy depletion, neuronal damage, and cell death

Oxidative stress

Compelling evidence has demonstrated the significantly increased oxidative stress in AD brains [37]. The imbalance between free radical production and antioxidant defenses in AD induces excessive oxidative stress in the AD brains [37]. As the “powerhouses of the cell,” mitochondria are also the primary intracellular source of oxygen radicals under both physiological and pathological conditions [22, 23]. As shown in Fig. 4, under physiological conditions, a small number of electrons leak out of the electron transport chain (ETC) and are transferred to oxygen to produce reactive oxygen species (ROS) [38]. The ROS generated serves as second messengers in mediating several critical intracellular pathways [39, 40]. In pathological situations, mitochondrial dysfunction and the impaired mitochondrial complex activity induce significantly increased ROS production. This leads to deleterious effects and a vicious cycle causing mitochondrial damage, energy depletion, neuronal damage, and cell death (Fig. 4) [41, 42]. Several studies demonstrate the increase in oxidative damage to DNA, proteins (protein carbonyl), and lipids (lipid peroxidation), which contribute to the initiation and progression of AD [43,44,45].

Neuroinflammation, polarization of glial cells, and microglial phagocytosis

Neuroinflammation triggered by pathological molecules including Aβ, tau, and damage-associated molecular patterns (DAMPs) in AD has been well demonstrated [44, 46]. In the early stages of AD, the deposition of Aβ acutely initiates the activation of microglia to remove Aβ through phagocytosis [47]. During this stage, the activation of microglia works as a protective response against AD [48]. However, microglial phagocytosis fails to remove amyloid plaques, and the increasing microglial activation releases an array of pro-inflammatory mediators that contribute to AD’s progression [49]. As shown in Fig. 5, the activated microglia can be divided into pro-inflammatory subtype M1 and M2 anti-inflammatory phenotype [50]. The M1 phenotype produces pro-inflammatory cytokines (i.e., IL-1β, TNF-α, IL-6, and IL-12), exacerbating AD progression. However, the M2 phenotype release anti-inflammatory cytokines (i.e., IL-4, IL-10, IL-13, and TGF-β), providing neuroprotective effects in AD [51, 52]. Furthermore, the M1 phenotype display relatively poor phagocytosis of Aβ, and the M2 phenotype shows elevated phagocytosis [53, 54]. Taken together, in the early stage of AD, the quiescent microglia are polarized into the M2 phenotype to confer a neuroprotective effect by releasing anti-inflammatory and neurotrophic factors. At the late stage, the microglia are polarized into the M1 phenotype, inducing neuronal loss and exacerbating AD progression by releasing pro-inflammatory cytokines and ROS (Fig. 5) [51]. Recently, a study using single-cell RNA-seq in the AD animal models identified a novel microglia type called disease-associated microglia (DAM), which represent a distinctive microglia states detected in AD but not in the wild type brains. The DAM coexists with the homeostatic microglia and infiltrating monocytes. Similarly, the transition between the homeostatic microglia to the DAM subtype is consistent with the direction of AD progression [55, 56].

Fig. 5
figure 5

Activation and polarization of microglia in AD. The M1 phenotype produces pro-inflammatory cytokines (i.e., IL-1β, TNF-α, NFκB, and IL-6), exacerbating AD progression. However, the M2 phenotype release anti-inflammatory cytokines (i.e., IL-4, IL-10, and IL-13), providing neuroprotective effects in AD. Similar to microglia, astrocytes are classified into A1 neurotoxic and A2 neurotrophic/neuroprotective phenotypes. In the early stage of AD, quiescent glial cells are activated to the A2 and M2 phenotypes and transformed into A1 and M1 phenotypes at the late stage of AD transformation

Similar to microglia, the activation of astrocyte, another sub-type of glial cells in the central nervous system, is also found near the sites of amyloid plaques [57]. In postmortem tissues from both AD patients and animal models, astrogliosis is observed and correlated with cognitive decline in AD [58,59,60]. According to recent studies, astrocytes are classified into A1 neurotoxic phenotype and A2 neurotrophic/neuroprotective phenotype [61, 62]. A1 phenotype can be activated by factors and fragmented mitochondria released from microglia to trigger and exacerbate neuroinflammation in AD [63, 64]. In contrast, the A2 phenotype is proposed to be a reparative and neuroprotective astrocyte phenotype in the brain [65]. According to previous studies, the phenotypical switch from the A1 phenotype to the A2 phenotype ameliorates AD pathology [66, 67].

A previous study showed that young microglia could restore the capacity of amyloid plaque clearance of aged microglia [68], and approaches that trigger the recruitment of microglia around amyloid plaques display a potential effect on attenuating AD pathology [69, 70]. Recently, the crosstalk between astrocytes and microglia provides a novel mechanism for the microglial recruitment around amyloid plaques [71]. In both the humans and mice, the astrocytic interleukin-3 (IL-3) can target the microglial IL-3 receptor (IL-3R) to induce the recruitment of microglia and enhance the ability to clear Aβ and tau, suggesting the astrocytic IL-3 is a crucial mediator of microglial recruitment and a potential target for the regulation of microglial phagocytosis [71].

Challenges for AD studies

Currently, no preventive or curative treatment is established to be safe and efficacious for AD [72]. Almost all molecular targeted pharmacologic therapies developed to treat or slow down AD have failed in clinical trials [73]. Although scientific advances in the past few decades have expanded our understanding of AD’s cellular and molecular basis, the exact mechanisms of AD generation remain to be fully unveiled [74]. Additionally, for AD patients, the pathophysiological changes of AD precede the clinical symptoms for many years [74]. For example, prominent deposition of amyloid plaques has already been displayed in the brain tissue before memory loss, which is difficult to be reversed [75]. Therefore, finding early detection biomarkers will be one of the primary research focuses of AD in future studies [74]. In addition, currently, nearly all the pharmacologic therapies in AD are based on the “lock-and-key” model, and the discovery of drugs focuses only on a single target (e.g., Aβ or tau) [76]. However, AD develops from various factors [77]. Therefore, further studies focusing on multiple targets may help facilitate AD treatment and prevention [76]. Moreover, experimental animal models are essential for better understanding of AD pathogenesis and progression and performing a preclinical assessment of the potential novel therapeutics [78]. To date, transgenic mice that express mutant forms of human APP, tau, and/or presenilin-1 (PS1) are considered as the “gold standard” animal models of AD [79]. However, these transgenic mice only have limited aspects of AD pathologies of humans, which hampers the successful transmission of animal experiments to clinical trials [78]. Therefore, choosing an appropriate animal model is essential for future AD studies.

PBM therapy and parameters of intervention

PBM therapy, also known as low-level laser (light) therapy, is a non-invasive photoceutical approach involving the application of relatively low levels of visible (wavelength between 400 and 720) or near-infrared light (wavelength between ~700 and 100 nm) on biological tissues to improve healing, relieve inflammation and pain, and preserve tissue function [22, 80]. As a non-invasive treatment method, PBM therapy describes the application of the low-level laser (light) directly to a specific region of interest on the body to modulate various biological processes [6]. It was first discovered by Endre Mester, a Hungarian physician working with wound healing and hair regrowth in 1967 [81, 82]. He found quicker hair growth and wound healing in the rats with low-level laser treatment [81, 82], which was the first study that found the beneficial application of low-level laser light on biological processes [83]. Since then, an increasing paper has described this treatment as “laser biostimulation,” “low-level laser (light) therapy (LLLT),” and now “PBM therapy” [84,85,86]. Although PBM therapy was initially applied to promote hair regrowth and wound healing [81, 82], the beneficial effects of PBM on relieving pain and inflammation and promoting muscle recovery have been widely studied [87,88,89,90]. In the past decades, the potential role of PBM therapy in the treatment of brain disorders has garnered increasing attention [7, 8, 91, 92]. Recent studies provide considerable evidence of PBM’s promising therapeutic potential in AD treatment [7, 69, 70].

Multiple parameters simultaneously affect the efficiency of PBM therapy, including wavelengths, intensities, durations, target area, and operation mode. Currently, the most widely used wavelength is red (~600–700 nm) and near-infrared wavelengths (~780–1100 nm) [93]. For the PBM therapy with wavelength within 600–1100, the primary target is the cytochrome c oxidase (CCO), the unite IV of the mitochondrial respiratory chain [22]. Additionally, evidence supports that blue and green light with shorter wavelengths within 450–570 nm can also confer PBM effects [94]. The effects may rely on intracellular calcium and light-gated ion channels [94]. However, the low transmission tissue limits the application of blue or green light within 450–570 nm [94].

Another critical parameter of PBM is the dose of the light source. The treatment doses of PBM depend on the intensities, duration, and PBM targeted area [93, 95, 96]. Currently, there is no consensus about treatment doses of PBM therapy. Except for the efficiency of the PBM therapy, the primary concern for the treatment dose of PBM is the thermal effects [97]. The intensities that produce the unacceptable thermal impact of the tissue are ~300 mW/cm2 at 600 to 700 nm, about 750 mW/cm2 at 800 to 900 nm, and 100 mW/cm2 at 400–500 nm [96]. Furthermore, the exposure duration is also critical for the efficiency of PBM therapy [96, 98]. Studies showed that even a few minutes of PBM therapy could cause biological changes [99, 100]. However, the best exposure duration also relies on other parameters used in the PBM application [96].

Aside from these parameters, the mode of operation of PBM can either be a pulsed wave or a continuous wave [101]. Both pulsed and continuous wave PBM display beneficial effects in AD [7, 69, 70, 102]. The pulsed wave PBM refers to the PBM effect induced by the light source in pulses of some duration at some repetition rate [103]. The most widely studied frequency is 10 Hz or 40 Hz in AD [69, 70]. Although different operation mode of PBM has similar effects, the underlying mechanism differs, which will be discussed in the following section.

PBM therapy for AD

Improves behavioral results and reduces amyloid plaques and neurofibrillary tangles

Intriguingly, increasing human and animal studies suggest that PBM treatment is a promising potential therapy in AD [69, 70, 104,105,106,107]. Progressive memory impairment is one of the primary signs of AD and is usually the first and common complaint for the patient to seek diagnosis [7, 108, 109]. As a hallmark of AD, memory impairment/loss is one of the most common clinically relevant markers to assess the effect of a potential treatment [110]. In a previous study, the learning and memory deficits were significantly improved by PBM at 40 Hz (light-emitting diode) in the AD mouse model [70]. The non-invasive 40-Hz light also reduced the levels of Aβ1-40 and Aβ1-42, regulated microglia’s morphological transformation, and improved both the phagocytosis and migration/cell adhesion-related genes in the cortex and hippocampus, which helped the improvement of cognitive function [70]. In a recent study, PBM with 1070-nm pulsed-wave light at 10 Hz could also improve learning and memory impairments after PBM treatment in AD mice [69]. Moreover, the AD animals with PBM therapy displayed reduced Aβ burden and improved Aβ clearance by regulating microglia and angiogenesis [69]. Another study found that PBM could promote the permeability of BBB, which results in increased Aβ leakage followed by further activation of the lymphatic clearance of Aβ [111]. As shown in Supplementary Table 1, the PBM has been applied to the frontal cortex, temporal regions, base of the skull, wrist, nose cavity, abdomen, and forehead bilaterally or generated gamma entrainment using sensory stimuli through the eyes in human clinical trials. Currently, most PBM-associated clinical trials are ongoing or terminated due to the coronavirus. Therefore, whether PBM can reduce the amyloid plaques in the whole brain is unclear. However, according to previous animal studies, the PBM therapy can alleviate at least amyloid plaques in the cortex and hippocampus [70, 112]. These findings added more evidence to the therapeutic effect of pulsed-mode PBM.

In addition to PBM with pulsed wave light, several studies identified the beneficial role of PBM with continuous wave light [7, 102]. In an Aβ-induced AD rat model, PBM treatment with the continuous-wave laser diode at 808 nm for 5 days protected against Aβ-induced cell toxicity and long-term spatial and object recognition memory [102]. In addition, the 5-day PBM treatment alleviated the hyperphosphorylated tau (PHF1) protein expression and neuronal apoptosis [102]. These findings were consistent with another research that found that PBM treatment with the continuous-wave laser at 632.8 nm suppressed neuronal loss and dendritic atrophy in the APP/PS1 double-transgenic AD mouse model [113]. Furthermore, anxious-depressive-like behavior has been detected and recognized as an early sign of AD pathogenesis [7]. Growing evidence implied that early treatment of anxious-depressive-like behavior could lower the risk of developing AD [7, 114]. The beneficial effect of PBM treatment in alleviating depression and anxiety-like behaviors has been widely reported [92, 115,116,117]. Intriguingly, PBM treatment with a continuous-wave laser could attenuate anxious-depressive-like behavior and protect against neuronal damage and apoptosis in the AD rat animal model, supporting the potential role of PBM therapy in preventing or slowing down the progression of AD [7].

Moreover, the beneficial effect of PBM on AD has also been identified by numerous clinical trials [104,105,106,107]. Using an 810-nm, 10-Hz pulsed LED light source PBM therapy, AD patients with moderate-to-severe cognitive impairment were assigned to receive 12-week PBM treatment. Results showed that 12 weeks of active PBM treatment significantly improved AD patients’ cognitive function and reduced anxiety [107]. Another clinical trial with continuous-wave near-infrared PBM therapy concluded that in PBM treatment patients displayed better cerebral perfusion and resting-state functional connectivity and a significantly improved cognitive and behavioral function [105]. Moreover, a case report further supported these previous observations [118]. In this case report, patients diagnosed with both cognitive decline and olfactory dysfunction received a mixture of continuous-wave mode red (635 nm), near-infrared light (NIR) LEDs (810 nm), and 10-Hz pulsed wave mode NIR (810 nm) PBM therapy. After PBM therapy, significant improvements were detected in the Montreal Cognitive Assessment and Working Memory Questionnaire [118]. As shown in Supplementary Table 1, an increasing number of clinical trials are testing non-invasive therapy in AD. Among these clinical trials, a clinical trial involving gamma entrainment therapies (also known as gamma entrainment using sensory stimuli, or GENUS) based on previous studies has entered phase II clinical trials in AD patients. Previous studies found that gamma entrainment therapies alleviate cognitive deficits and improve the clearance of amyloid plaques by recruiting neuronal and glial responses, shifting neurons to a less degenerative state, releasing neuroprotective factors, enhancing synaptic function, alleviating neuroinflammation, and reducing DNA damage-associated cytotoxicity in neurons [70, 112]. In these studies, 40-Hz pulsed light or 40-Hz blue light with 40-Hz auditory stimulation was employed to produce GENUS and exhibited neuroprotective effects in the AD mouse model, suggesting an alternate form of PBM in treating neurodegenerative diseases. Overall, these findings support the potential therapeutic application of PBM therapy in improving cognitive impairment and reducing anxious-depressive-like behaviors of AD.

Preserves mitochondrial function and structure

As mentioned previously, mitochondrial dysfunction and aberrant mitochondrial fragmentation are involved in the occurrence and development of AD and are recognized as common features of neurodegenerative disease [22]. It is widely accepted that mitochondrial cytochrome c oxidase (CCO, complex IV of the respiratory chain) is the primary action site of PBM therapy [8, 22]. Therefore, a large body of evidence suggests that the beneficial effect of PBM therapy on AD is mainly due to the regulation of mitochondrial function and mitochondria-related processes [8, 22]. The primary mechanism underlying PBM’s regulation of mitochondria is involved in the modulation of CCO activity [119,120,121,122]. Nitric oxide (NO), a molecule that binds non-covalently to the heme iron and copper centers of CCO to inhibit the activity, is the most important medium in this process [119,120,121,122]. PBM therapy enhances CCO activity by photodissociating NO from CCO, thereby reversing the inhibition of the electron transport chain due to excessive NO binding (Fig. 6) [123, 124]. The effect of PBM on CCO indicates that PBM treatment is a potential intervention for promoting mitochondrial function in AD [123].

Fig. 6
figure 6

PBM promotes CCO activity and increases ATP production. Nitric oxide (NO) inhibits CCO activity by non-covalently binding with CCO. PBM treatment leads to the dissociation of NO from CCO, causing the increased activity of this complex and ATP production

Besides the direct effect on mitochondrial CCO, PBM therapy regulates mitochondrial dynamic and fragmentation [125, 126]. The balance of mitochondria fission and fusion is crucial for the normal function of mitochondria and the maintenance of mitochondrial morphology [22, 127]. Aberrant or increased mitochondrial fission in AD leads to increased mitochondrial fragmentation and neuronal death [128]. Intriguingly, several previous studies have demonstrated that PBM treatment preserved the dynamic equilibrium between mitochondrial fusion and fission in various brain diseases such as global cerebral ischemia, neonatal hypoxic ischemia, AD, and Parkinson’s disease [8, 125, 129, 130]. In an Aβ1-42-induced AD rat model, PBM treatment with continuous-wave low-level diode laser significantly alleviated excessive mitochondrial fission-induced mitochondrial fragmentation by promoting mitochondrial fusion-related proteins (OPA1 and MFN1) and inhibiting mitochondrial fission-related protein (Drp1, Fis1, Mff, and Mief) [102]. Similar regulation of these proteins was also observed in other brain disease models [8, 22, 119]. Taken together, PBM, a mitochondria-targeted therapy, demonstrated its therapeutic potential in AD treatment by regulating the mitochondrial structure and mitochondrial function.

Regulates glial cells and exerts anti-inflammatory effects

Emerging evidence from previous studies demonstrated the beneficial role of PBM therapy in regulating glial cells and neuroinflammation [8, 69, 70, 121, 131]. In a previous study, 40-Hz light flicker PBM therapy recruited microglia around amyloid plaques and improved microglial phagocytosis and migration/cell adhesion-related genes in multiple mouse models [70]. The modification of microglia after PBM treatment triggers microglia to increase Aβ uptake [70], indicating the potential role of PBM treatment in improving microglial phagocytosis. Recently, results from another study further our understanding of PBM treatment on the regulation of glial cells [69]. As reported in their work, PBM treatment with 1070-nm pulsed-wave light at 10 Hz can also reduce cerebral Aβ burden by promoting the activation of microglia (e.g., increased cell body, reduced number and length of branches) and microglial phagocytosis [69]. Notably, they found a decreased pro-inflammatory M1 phenotype and the increased M2 anti-inflammatory phenotype after PBM treatment [69]. These findings demonstrate that PBM can inhibit neuroinflammation by promoting the transformation of microglia from a neurotoxic to a neuroprotective phenotype in AD [69]. Anti-inflammatory effects were also found in PBM therapy using continuous-wave lasers [102]. In the Aβ1-42-induced AD rat model, Aβ injection into the hippocampus of rats led to the increased release of pro-inflammatory cytokines (i.e., IL-1β, IL-5, and TNF-α), mitochondrial dysfunction, demyelination, and axonal damage of neurons [102, 132, 133]. Interestingly, PBM treatment with continuous-wave low-level diode laser significantly suppressed Aβ-induced neuroinflammation and protected against Aβ-induced neuronal injury and neurodegeneration [102]. However, in this AD rat model, PBM treatment with continuous-wave lasers significantly suppressed Aβ-induced reactive gliosis [102], which is different from the pulsed-wave PBM’s promoting the activation of glial cells [102]. One of the explanations for the difference may be due to the animal model. Aβ-induced AD-like rat model is an acute AD rat model, wherein Aβ induces reactive gliosis rapidly. However, in the transgenic mouse model, glial cells are activated progressively. PBM inhibits glial cells’ acute activation to protect against inflammatory response-induced neuronal damage in the acute AD animal models but promotes the activation and polarization of glial cells to a neuroprotective phenotype in the progressively developed AD models [51, 102].

Inhibits oxidative stress and oxidative damage

The anti-oxidative effect of PBM has been widely studied in the skeletal muscle after physical exercise [134,135,136]. Oxidative stress is also implicated in AD’s pathogenesis and progression [37, 137]. The significantly elevated levels of 4-hydroxyhexenal (4HHE), a lipid peroxidation marker, have been found in animal models and in vitro cell cultures [44, 138]. Moreover, the level of 8-hydroxydeoxyguanosine (8-OHdG), a widely used DNA oxidative marker, was a threefold increase in the postmortem brain tissue of AD patients compared with age-matched controls [139]. Similar results were found in AD-like animal models [44, 140]. Furthermore, the oxidative damage of protein induced by excessive free radicals was detected in sporadic AD rat and transgenic AD models [7, 44, 141]. Additionally, several enzymes critical to neuron and glial functions are prone to oxidative damage and decrease in AD [37]. For example, the enzymes susceptible to oxidative stress, the glutamine synthetase and creatine kinase, are significantly reduced in AD brains, inducing decreased glutamate concentrations and excitotoxicity enhancement [37, 141]. Moreover, oxidative stress impaired creatine kinase activity and caused reduced energy metabolism in AD [37, 141]. Interestingly, PBM with different light sources and parameters shows its significant anti-oxidative effect in neuronal cell culture and AD brains. PBM treatment with 660-nm continuous-wave LED at 20 mW/cm2 protects against neuronal cell death by reducing H2O2-induced oxidative stress in vitro study [142], wherein the improved antioxidant enzyme and redox homeostasis are considered as the key mechanism underlying this protection [142]. Further evidence from two transgenic mouse models (APP/PS1 and K369I tau transgenic model) supports the anti-oxidative effect of 670-nm near-infrared light [143]. After receiving 90-s PBM treatment for 5 days per week, the AD animal displayed a significant reduction in neurofibrillary tangles, hyperphosphorylated tau, and oxidative stress markers (4-HNE and 8-OHdG) in the cortex and hippocampus [143].

The anti-oxidative effect of PBM is closely related to the preservation of mitochondrial function [22]. As mentioned above, the primary action sit of PBM therapy is CCO [8, 22]. A short burst of ROS is generated when NO is photodissociated from CCO after PBM treatment [8, 22]. The temporary and relatively modest increase of ROS production leads to the activation of the NF-κB and PI3K/Akt pathway [125, 144, 145]. The activation of NF-κB will lead to the translocation from the cytoplasm to the nucleus. It induces more than 150 gene expressions, including genes associated with antioxidant activity and mitochondrial dynamics [22, 146]. Therefore, oxidative stress is significantly alleviated in the cells subjected to stress with PBM treatment [147]. This evidence explains how PBM therapy reduces clinical oxidative stress and protected neurons from death in various lesions and AD [147].

Challenges of PBM therapy in AD

Although the beneficial effects of PBM therapy have been found in different tissues and widely studied in brain disease, challenges remain in the clinical application of PBM on AD patients [83]. First, there is no agreement on the parameters and protocols of PBM therapy in the clinical application of AD [96]. As mentioned previously, several light source parameters are involved in the effects of PBM therapy. The prevailing use of a wide variety of light sources, the illumination parameters (e.g., wavelength, power density, pulse structure, fluence), and treatment protocols (a single application of light or multiple doses) induce significant variations in the study design [83, 98]. The disagreement on light/laser parameters and the variations in study designs led to many negative results in clinical trials and posed some controversy in PBM’s study and application [83]. Second, although CCO has been widely accepted as the primary target of PBM, there is still conflicting data doubting whether it is the primary acceptor [148, 149]. For example, a study found that PBM could promote ATP production and increase cell proliferation in CCO knockout cells, suggesting that CCO may not be the primary or only target of PBM therapy [149]. Therefore, more studies are still needed for the precise mechanisms of PBM treatment in AD. Although increasing evidence supported the beneficial effect of pulsed-wave PBM therapy, the exact mechanisms of pulsed-wave PBM therapy in AD remain elusive [69, 70]. Finally, in animal studies and clinical trials, almost all studies performed PBM directly at the target tissue. However, increasing studies over the past several years demonstrated that the beneficial effects of PBM therapy are not limited to irradiated tissue [150, 151]. However, the indirect or remote PBM therapy mechanisms remain to be understood. More studies on the remote PBM therapy have far-reaching implications on the therapeutic application of PBM in AD as the brain is a difficult-to-irradiate organ [150, 151].

Transcranial magnetic stimulation and parameters of intervention

Transcranial magnetic stimulation (TMS) is another non-invasive approach developed to treat multiple neurodegenerative diseases such as AD [152, 153]. TMS relies on electromagnetic pulses to stimulate brain cells wherein the changing electric-induced magnetic field is applied to a target region of the brain [10, 154]. Generally, the apparatus used for TMS is composed of an electric pulse generator (stimulator) and a magnetic coil [155]. The electric pulse generator is connected to the magnetic coil, which produces a magnetic field delivering non-invasive magnetic pulses to a specific brain area at different intervals and frequencies [155]. The physical principles of TMS were first described by Michael Faraday, an English physicist, in 1881 [156]. Then the magnetic field targeting a specific brain was found to induce physiological and behavioral changes [156, 157]. After that, Anthony Barker and his team first introduced the TMS device and stimulated the brain’s neurons in the left cerebral motor strip to move the right hand, which was the first time the TMS was used to direct physical response and also a demonstration of TMS being capable of non-invasively stimulating a specific area of the brain without causing pain which is a case for electrical stimulation [158]. Currently, the repetitive transcranial magnetic stimulation (rTMS) delivering repetitive magnetic pulses has been adopted into clinical practice for the treatment of major depressive disorder (MDD) [159], and now when people mention the TMS therapy, it usually refers to rTMS [159]. The development of rTMS as an antidepressant therapy has been supported by multiple randomized controlled trials and published literature [160,161,162]. In the clinical trials, patients with rTMS treatment showed significant improvement in the depressive symptoms and higher rates of remission [160,161,162,163,164]. Therefore, since the US Food and Drug Administration (FDA) approved the first device for rTMS treatment of MDD in 2008, FDA has cleared 5 TMS devices for MDD treatment in the USA [160]. The safety and efficacy of rTMS in anti-depression drives people’s interest in other neurological diseases.

Similar to PBM, the effect of rTMS relies on several parameters, including interstimulus interval, stimulus intensity, number of stimuli, the interval between successive trains, stimulus duration, and delivery mode [165]. The long-lasting after-effect triggered by rTMS treatment depends on the combinations of different parameters [166]. In general, the operation of rTMS can be divided into “low or high frequency” and “conventional or patterned TMS” [165, 166]. Low-frequency stimulation and high-frequency refer to stimulation rates lower than 1 Hz and higher than 5 Hz, respectively [165, 166]. In most low-frequency rTMS studies, the low-frequency therapy induced an inhibitory effect on cortical excitability, and high-frequency rTMS led to an excitatory effect [166]. rTMS with 10 Hz and 20 Hz is the most commonly used frequency in AD clinical trials [166,167,168]. The conventional TMS refers to a single TMS pulse application in a regular rhythm, and the patterned rTMS refers to the application of TMS with short, high-frequency bursts with brief periods of no stimulation [166]. Increasing evidence supports the beneficial effects of theta-burst stimulation (TBS) on AD recently [152, 169]. TBS is a typically patterned rTMS, wherein short bursts of high-frequency pulses repeated at 5 Hz are delivered based on the natural theta rhythm in the hippocampus [152, 166]. In terms of delivery modes, the TBS can be further divided into continuous TBS (uninterrupted sequence) and intermittent TBS [166, 170]. As mentioned above, beyond stimulation frequency and delivery mode, stimulus intensity and duration are important factors influencing the outcome of rTMS. However, unfortunately, there is no consensus on the optimal stimulation protocols [165].

rTMS therapy for AD

Improves cognitive function of AD and decreases Aβ accumulation and tauopathy

Increasing evidence from clinical and animal studies supported rTMS as a promising treatment for mild and moderate AD [171,172,173]. At the early stages of AD, high-frequency rTMS treatment with 40 burst trains at 20 Hz was delivered to the head of a transgenic familial AD mouse model (5xFAD mice). After 14 consecutive days of rTMS treatment, AD animals with rTMS displayed significant improvement in long-term memory performance, suggesting the beneficial effects of rTMS with 20 Hz for memory impairment [171]. Similar to high-frequency rTMS treatment, relatively low-frequency rTMS treatment also plays a beneficial role in improving cognitive impairment [174]. Furthermore, rTMS at 5 Hz delivered to the head for 14 consecutive days significantly also enhanced the learning and memory of APP/PS1 mice, as evidenced by shortened escape latency and increased time in the target quadrant in the AD-like mouse model rTMS treatment [174]. Consistent with the behavioral results, rTMS at 5 Hz reduced neuronal Aβ accumulation and tau hyperphosphorylation, the hallmark pathologies in AD progress [174, 175]. The underlying mechanisms include reducing Apolipoprotein E (ApoE) expression and promoting autophagic flux following rTMS treatment, which is in line with a previous finding that rTMS promotes the BBB-mediated drainage efficiency of the brain clearance pathways in an AD animal model [171, 174].

Mild cognitive impairment (MCI) is one of the typical symptoms of AD [176]. In a scopolamine-induced AD-related MCI mouse model, both the low- and high-frequency rTMS (1 Hz and 10 Hz) were applied to the head of the rats for two sessions. AD mouse with rTMS treatment showed significantly improved cognitive function in a frequency-dependent manner, with the high-frequency rTMS exhibiting better improvement effects on cognitive function [176]. Similar results were found in the Aβ42 administration-induced AD mouse model [177]. Both the low-frequency (1 Hz) and high-frequency (20 Hz) rTMS were able to enhance spatial working memory in a frequency-dependent manner [177], suggesting rTMS therapy exists the optimal dosage, which needs more study. Notably, the effects of rTMS do not only rely on the frequency [178]. It also depends on the intensity of rTMS [178]. The low frequency of rTMS at high intensity appeared to be detrimental to cognitive function [178].

Clinical trials confirmed the effects of rTMS on the cognitive function of AD [175, 179]. For example, AD patients who received high-frequency rTMS at 20 Hz for 2 weeks showed improved auditory sentence comprehension, suggesting that rTMS could modulate short-and/or long-range cortical synaptic efficacy and connectivity, leading to more effective processing [175]. This effect is also found in another rTMS clinical trial with 20 Hz for 6 weeks, in which more measurements were performed to measure cognitive function, including Assessment Scale-cognitive subscale (ADAS-cog), Montreal Cognitive Assessment (MoCA), Mini-Mental State Examination (MMSE), and the World Health Organization-University of California-Los Angeles Auditory Verbal Learning Test (WHO-UCLA AVLT) [179]. In these tests, the MMSE, ADAS-cog, and WHO-UCLA AVLT scores in the rTMS group were significantly improved, suggesting that rTMS is also a promising therapy for enhancing cognitive function in AD patients [179].

Promotes synaptic plasticity and hippocampal neurogenesis

rTMS modulates neuronal processing by inducing the depolarization of neural cell membrane potentials under the magnetic field and affecting the related nerve loop activity with prolonged effects on neural activity [154, 180]. The regulation of synaptic plasticity is the most widely accepted mechanism of rTMS [178]. It modulates long-term potentiation/depression (LTP/LTD) of excitatory synaptic transmission and influences spatial cognition [178]. rTMS-induced LTP or LTD depends on the strength of Ca2+ internal flow and the intracellular Ca2+ level in the postsynaptic membrane, which is determined by the combinations of the rTMS’s parameters [181]. In both the vascular dementia animal model and normal aging model, low-frequency rTMS (1 Hz) at low intensity alleviates cognitive impairment by increasing the expressions of synaptic protein markers and activating brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB), the key mediators of neuronal and synaptic maturation [178, 182].

Besides the changes in synapses, neuronal loss is another of the hallmarks of AD [183]. Neurogenesis is abundant in healthy individuals [184]. However, the impaired adult hippocampal neurogenesis in AD patients exacerbates neuronal loss and contributes to AD progression [184]. Intriguingly, studies detected neurogenesis after rTMS treatment [178, 185,186,187]. In a focal cerebral ischemia animal model, rTMS treatment at both low frequency (1 Hz) and high frequency (20 Hz) was analyzed [185]. Notably, high-frequency rTMS at 20 Hz significantly improved neurogenesis in the ischemic striatum [185]. However, neurogenesis was not markedly elevated by rTMS at 1 Hz [185]. Consistent with these findings, the infarct volume was decreased, and functional recovery was enhanced after high-frequency rTMS treatment [185]. In contrast, the neurogenesis was suppressed in the chronic psychosocial stress animal model after 18 days of rTMS treatment at the same frequency [185]. The difference in the other parameters of rTMS, including the intensity and treatment duration, may explain the discrepancies between these two studies. Furthermore, the different animal models may be another possible reason for the different neurobiological effects [187]. A previous study confirmed one of the explanations that intensity may affect the neurobiological effects of rTMS [188]. In their study, rTMS with the same frequency but different intensities was applied on a depressive-like model [188], wherein only medium-intensity rTMS at 50 mT increased BDNF and neurogenesis [188]. Consistent with the findings in the brain injury model, rTMS with intermittent gamma burst stimulation (30–40 Hz) can promote neurogenesis and differentiation of newborn cells into mature neurons in the hippocampus [186]. Although discrepancies exist in the neurogenesis after rTMS treatment in different animal models, increasing evidence suggests rTMS with appropriate parameters can promote neurogenesis [189,190,191]. More studies on neurogenesis and synaptic plasticity after rTMS are still needed in AD.

Neurotransmitters contribute to the beneficial effects of rTMS

The regulation of neurotransmitters is another possible mechanism contributing to the beneficial effects of rTMS [154]. Anxious-depressive-like behavior has been recognized as an early sign of AD [7, 9, 192, 193]. Furthermore, increasing evidence indicated that treatment of depressive-like behavior could attenuate later cognitive deficits, thereby presenting a target to slow AD development [114]. Therefore, the regulation of neurotransmitters associated with anxious-depressive-like behavior and cognitive function contributes to the beneficial effects of rTMS stimulation [154].

Serotonin (5-HT) is an essential excitatory transmitter playing a critical role in neuropsychiatric disorders and memory loss [194, 195]. However, the cortical 5-HT and 5-HT receptor levels were reduced in the postmortem brain tissue of AD patients and patients with major depressive disorder [196,197,198]. Interestingly, high-frequency rTMS at 10 Hz applied to the left dorsolateral prefrontal cortex (DLPFC) significantly upregulates 5-HT content and 5-HT receptors [199,200,201,202]. Similar results are also found after treatment with low-frequency rTMS at 1 Hz [203, 204]. Furthermore, preclinical studies and clinical trials have demonstrated that dopamine contributes to the pathophysiology of depression and the preservation of cognitive function and dendritic spine structure [205,206,207]. Intriguingly, rTMS was found to increase dopamine release in the mesostriatal, mesolimbic, and striatal regions [154, 208].

Furthermore, gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain [209]. The dysfunction of the GABAergic system contributes to the pathophysiology of depression and cognitive impairment in humans [210, 211]. Significant reductions in GABA levels have been found in severe AD cases, which are involved in the psychological symptoms of AD [210]. Notably, 10-Hz rTMS led to elevated prefrontal cortex GABA in patients with major depressive disorder. In contrast, high-frequency (25 Hz) rTMS could reverse the increased GABA in PFC during aging [212, 213], suggesting rTMS can regulate GABA to a different level in different conditions and maintain the homeostasis of GABA level [212, 213].

Attenuates neuroinflammation and regulates glial cells

The anti-inflammatory effects of rTMS have been widely reported in various brain diseases, including Parkinson’s disease [214], focal cerebellar injury [215], ischemic stroke [216], depression [217], and anxiety [218]. Notably, the anti-inflammatory effects of rTMS also contribute to its beneficial role in AD [219]. rTMS at 20 Hz suppressed the overactivation of microglia and pro-inflammatory cytokine levels at the early stage of 5xFAD mice [219]. In AD, the increased release of pro-inflammatory cytokines, such as TNF-α, binds to the cell membrane receptors and induces the intracellular PI3K/Akt/NF-κB signaling pathway activation [219]. The increased NF-κB binds with DNA to further cause the release of pro-inflammatory in both the cortex and hippocampus of AD [219]. However, treatment with rTMS at 20 Hz significantly reduces the phosphorylation of Akt and the translocation of the p65 subunit of NF-κB into the nucleus, suggesting rTMS suppressed the neuroinflammation by inhibiting the excessive activation of PI3K/Akt/NF-κB signaling pathway [219]. Finally, the anti-inflammatory of 20-Hz rTMS significantly improves the neuronal environment and alleviates synaptic plasticity impairment in AD animals [219].

Although there is no direct evidence demonstrating the switch between pro-inflammatory M1 microglia and anti-inflammatory M2 microglia in AD after rTMS treatment, the transformation between M1 and M2 was detected in other brain disease models [10]. In our previous study, rTMS with a standard theta-burst stimulation paradigm (3 pulses of 50 Hz, repeated every 200 ms) were applied to an ischemic stroke model [10]. We found that rTMS effectively induce a switch between M1 and M2 phenotype [10]. Interestingly, a shift in astrocytic A1/A2 phenotype was also detected in this stroke model after rTMS therapy [10]. The release of pro-inflammatory cytokines characterizes M1 and A1 phenotypes, and M2 and A2 are associated with increased production of anti-inflammatory cytokines, the removal of cellular debris, and tissue repair [8]. Intriguingly, consistent with the polarization of glial cells, anti-inflammatory cytokines are increased, and the pro-inflammatory cytokines are decreased [8]. Additionally, the transformation of astrocyte from neurotoxic A1 phenotype to neuroprotective A2 phenotype was also found in ischemic stroke treated with rTMS stimulation at 10 Hz [216].

Moreover, the anti-inflammatory effect of rTMS was also confirmed by in vitro studies [215, 220]. In the primary cortical astrocyte culture, low-frequency and high-frequency stimulation alleviated oxygen-glucose deprivation (OGD) induced-neurotoxic A1 polarization [216]. Similarly, the pro-inflammatory mediator/anti-inflammatory cytokine concentration was also decreased/increased, consistent with in vivo study [216]. Aberrant increased calcium signaling contributes to excessive astrocytic activation and is a good indicator of disease severity/state in neurological disorders [220, 221]. Interestingly, rTMS leads to a significant down-regulation of calcium signal-related genes and inflammatory molecules, suggesting that rTMS has a potential role in regulating the AD inflammatory response [220].

Inhibits oxidative stress and preserves mitochondrial function

A large body of evidence demonstrates that oxidative stress is a prominent early event in AD and a promising biomarker for predicting treatment effects [222]. Interestingly, rTMS display its anti-oxidative effects in multiple brain diseases [223, 224]. For example, multiple sclerosis patients who received long-term treatment with 1-Hz rTMS showed significant improvement in various levels of psychometric evaluation and blood analysis [225]. Notably, there is a marked reduction of oxidative stress in the plasma [225]. Moreover, as an approved first-line treatment for depression in many countries [226, 227], rTMS also show its anti-oxidative effect on depression [228]. Although the relationship between reduced oxidative stress and improvement in depression was not established, patients with medication-resistant major depression displayed a significantly decreased level of oxidative stress marker in the blood sample after rTMS treatment [228].

Additionally, effective rTMS at a high frequency can alleviate oxidative stress [229]. In a 3-nitropropionic acid-induced oxidative stress model, the stress rats receiving 60-Hz rTMS displayed significantly decreased oxidative products and improved anti-oxidative enzyme activity in cortical synaptosomes [229]. Interestingly, rTMS did not affect the levels of these oxidative products and enzyme activity in the normal cortical synaptosomes, suggesting that rTMS is only involved in maintaining cellular redox homeostasis in pathological conditions [229]. Similarly, after ischemic stroke, rTMS treatment at 50 Hz significantly suppressed NADPH oxidase activation and superoxide levels in the peri-infarct cortical proteins [224]. It was also accompanied by markedly reduced expression of oxidative neuronal damage markers, including MDA lipid peroxidation marker (MDA) and DNA damage markers (e.g., p-H2A.X Ser139 and 8-OHdG) [224].

The oxidative damage is closely associated with mitochondrial function [22], and the disruption of the mitochondrial membrane was found in various brain diseases [22]. Intriguingly, the disrupted mitochondrial membrane potential is alleviated by rTMS at 50 Hz in ischemic stroke, suggesting the potential role of rTMS in modulating mitochondrial function [10]. Although evidence suggests the beneficial role of rTMS in multiple diseases through its anti-oxidative stress, studies in AD are still rare. In a clinical study, AD patients receiving 20-Hz rTMS targeting the left parietal region showed a significant reduction in blood oxidative damage markers in AD patients [223]. Consistent with the changes in oxidative stress, the cognitive function and the network connection between the hippocampus and the left parietal lobe were significantly improved, suggesting that the antioxidant effects of rTMS contribute to the beneficial effects of rTMS in the treatment of AD [223].

Alleviates neuronal apoptosis and provides neuroprotection

Due to the improvement of the above-mentioned multiple factors, rTMS has powerful anti-apoptotic and neuroprotective effects [230]. The Nissl bodies of the neurons in depressed rats were significantly reduced, and the cells were atrophied considerably or degenerated with irregular morphology, suggesting neuronal damage or apoptosis in depressed rats [230]. In contrast, more extensive and granular Nissl bodies were detected in the hippocampal neurons with well-defined cell boundaries after rTMS treatment, suggesting the anti-apoptotic and neuroprotective effect of rTMS in the depression [230]. Furthermore, the expression of the apoptosis regulator BAX was significantly suppressed by rTMS, which further confirmed the anti-apoptotic effect of rTMS in depression [230]. The anti-apoptotic and neuroprotective effect of rTMS is also detected in other disease models with higher rTMS frequency [224]. rTMS treatment at 50 Hz reduced the neuronal apoptosis in the ischemic stroke, supported by significantly suppressing the intrinsic apoptotic pathway in the peri-infarct area [224].

In AD mice, Aβ injection increased neuronal apoptosis, cleaved caspase-3, and Bax levels and decreased levels of Bcl-2 [231]. In contrast, after the treatments with rTMS at 1 Hz, the neuronal apoptosis was significantly suppressed, and apoptosis-related protein expressions were reversed, suggesting the anti-apoptosis effects of rTMS in AD [231]. Furthermore, besides the anti-apoptotic effects, rTMS enhanced the production of the brain-derived neurotrophic factor and nerve growth factor in the brain tissue of AD mice [231]. Both neurotrophic factors are critical molecules in plastic changes associated with learning and memory [231]. These findings suggest that rTMS exerts neurogenic and neuroprotective effects [177, 231].

Challenges of rTMS therapy in AD

Although rTMS has emerged as a promising approach to slowing down the AD progression or treatment, the evidence regarding long-term efficacy and exact underlying mechanisms is still limited [153]. Additionally, although, as noted earlier, several studies reported positive rTMS effects in AD [179, 232], more advanced clinical trials are still needed to find the therapeutic window for AD treatment [153]. For example, similar to other interventions [44], rTMS might be more effective at the early stage of the disease. Therefore, the rTMS should be applied before the neuronal loss and extensive Aβ deposition in AD [153]. Furthermore, most of the rTMS protocols used to treat AD are similar to those applied in the treatment of medication-resistant depression [153]. There is no agreement on the parameters and protocols of rTMS therapy at which a medication appears to be effective in the clinical application of AD [153]. Moreover, although studies have found that rTMS could correct or blunt the impaired LTP-like plasticity and exert a neuroprotective effect, there is no widely accepted target for rTMS therapy [153, 233]. Furthermore, the modulation of rTMS on mitochondria and glial cells’ transformation has been detected in other brain injuries, but no evidence is found in AD [10]. Finally, although rare (<1% overall), seizures are a potential side effect of TMS treatment [234]. Therefore, the safety of rTMS treatment, how to alleviate this side effect, and determining if there are other possible adverse events of TMS treatment need to be clarified [234].

Transcranial direct current stimulation (tDCS) and parameters of intervention

Transcranial direct current stimulation (tDCS) is a painless, non-invasive brain stimulation therapy that uses direct current to stimulate specific parts of the brain and produce facilitatory or inhibitory effects [235]. tDCS is typically applied using a constant, low-level current that passes through two electrodes positioned on the scalp to modulate neuronal activity [235]. The tDCS can be divided into two types of stimulation: the positive anodal current stimulation and the negative cathodal current stimulation [235]. The positive anodal current excites neuronal activity in the cortical region under the target electrode, whereas the negative cathodal current reduces or inhibits neuronal activity [235]. As a user-friendly, relatively cheap, and tolerable device, the beneficial effects of tDCS have been studied in various brain diseases, including depression [236], anxiety [237], Parkinson’s disease [238], traumatic brain injury [239], and AD [240]. Although tDCS is not an FDA-approved therapy for AD currently, increasing evidence supports the potential use of tDCS in AD prevention or treatment [241, 242].

tDCS therapy for AD

Improves cognitive function and reduces the deposition of Aβ

In recent years, emerging studies support the promising effect of tDCS in improving cognitive function in AD animals and patients [240, 243]. Repetitive anodal tDCS exhibited distinct neuroprotective effects on the AD brain [243]. To go into greater detail, after 10-day tDCS therapy, the animal showed significantly improved cognitive function and displayed a long-term after-effect that persisted for 2 months [243]. The effects of tDCS on cognitive function have also been investigated in healthy individuals and AD patients [244]. For example, the anodal tDCS could regulate cortical excitability by enhancing depolarization, whereas cathodal tDCS reduces cortical excitability by promoting neuronal hyperpolarization [244]. In human studies, anodal tDCS is applied to stimulate different cortical areas [245, 246]. The anodal tDCS placed on the left prefrontal cortex was able to improve the working memory performance of the healthy individuals [246], which was confirmed by another study in which the tDCS was placed over the left dorsolateral prefrontal cortex (DLPFC) [247]. Interestingly, when the tDCS was placed over the left DLPFC, the improved working memory performance could last for around 30 min, suggesting the potential use of tDCS in individuals with cognitive deficits [247].

In AD patients, anodal tDCS improve visual recognition memory [248], word recognition memory [249], face-name association memory [249], and cognitive function [242]. In a clinical experiment, both the short-term (10 days) and long-term tDCS (10 days/month for 8 months) were applied to AD patients [250]. Results showed that both the short-term and long-term anodal tDCS intervention effectively slowed down the progression of AD and preserved the neuropsychological performance [250]. This clinical experiment also suggests that if an anodal tDCS intervention is effective in the short term by maintaining the neurocognitive function, this beneficial effect can be prolonged over 8 months [250].

Furthermore, tDCS can reduce the formation of amyloid plaques [251]. For example, in an early-stage AD mouse model, 10-day tDCS was applied to the skull over the frontal cortex of the mice [251]. The mice that received tDCS had significantly improved spatial learning and memory, although no improvement in the recognition memory was found [251]. Notably, the Aβ42 deposition was significantly reduced in the tDCS-treated group, which provided direct evidence for alleviating the specific pathological change of AD tDCS [251]. In preclinical AD, the tDCS exhibited its beneficial role in alleviating neurovascular unit dysfunction and reducing Aβ plaques, indicating that the effect of tDCS on Aβ deposition may partly depend on BBB-mediated clearance of Aβ [252].

Improves cerebral blood flow

Previous studies found brain hypoperfusion or decreased cerebral blood flow in individuals with mild to moderate AD [253, 254]. Moreover, the compromised cerebral blood flow has been recognized as one of the early events of AD, predicting the progression of the disease and closely correlating with cognitive decline [255, 256]. Improving cerebral blood flow could alleviate the cognitive deficits in the AD animal model and human patients [257,258,259]. Intriguingly, tDCS show its ability to improve cerebral blood flow in healthy individuals during stimulation [260]. However, the cerebral blood flow returned to baseline when the tDCS was removed [260]. Moreover, cerebral blood flow change exhibited a linear relationship with tDCS intensity [260], suggesting that the regulation of tDCS on cerebral blood flow is intensity-dependent.

Unlike anodal tDCS, the cathodal tDCS induced a reduced cerebral blood flow during stimulation and a continuous decrease compared to baseline [260]. Similar to the human finding, the anodal tDCS also increased cerebral blood flow in rats [261]. However, unlike the human study noted previously, the effect of tDCS can last after tDCS therapy, suggesting that tDCS may have long-lasting after-effects in regulating cerebral blood flow [261]. Consistent with this result, there is an increased oxygen delivery in the vicinity of the anode after tDCS with after-effects lasting for several minutes after stimulation [262].

Alleviates impaired synaptic plasticity

Synaptic plasticity is the ability of synapses to change their strength or efficacy of synaptic transmission in response to synaptic activity changes [263]. In AD, impaired synaptic plasticity is an early event involved in the cognitive deficits [264]. Targeting the impaired synaptic plasticity with the brain stimulation techniques has been considered a powerful approach for treating AD [265]. Interestingly, the tDCS is one such method [266]. tDCS could induce membrane polarization and polarity-specific shifts of cortical excitability during and after stimulation and modulate the conductance of sodium and calcium channels [266]. These changes are involved in neuroplastic changes and provide a possible mechanism underlying long-lasting after-effects [266].

Moreover, the long-lasting after-effects of tDCS depended on NMDA receptor-associated neuroplastic changes [266]. The changes in NMDA receptor and intracellular Ca2+ uptake are one of the primary physiological bases of tDCS [267]. As mentioned previously, anodal tDCS enhances cortical activity, while cathodal stimulation reduces cortical excitability [244]. When the NMDA receptor was activated by anodal tDCS, the intracellular Ca2+ within the postsynaptic neuron was significantly increased [267, 268]. The effect of tDCS on synaptic plasticity depends on the degree of NMDA receptor activation [267, 268]. A slight increase of the intracellular Ca2+ in the postsynaptic neuron will lead to LTD-like changes [267, 268]. On the other hand, a moderate rise in Ca2+ does not affect the synaptic plasticity, and a more significant increase induces LTP-like changes [267, 268].

Regulates neurotransmitter systems

As mentioned previously, the AD rat model improved long-lasting cognitive function and memory performance after tDCS therapy [243]. The molecular study found decreased choline acetyltransferase (ChAT) levels in AD rats. In contrast, a notable increase of ChAT in the tDCS group [243] suggests that neurotransmitters’ changes may be involved in the effects of tDCS. ChAT is a transferase enzyme that facilitates the synthesis of the neurotransmitter acetylcholine (ACh) [269]. It is assumed that ChAT plays a crucial role in learning and memory [269]. In the AD rat model, Aβ-induced neurotoxicity led to the cholinergic system’s disruption and decreased ACh level [270]. Therefore, the increased expression of ChAT after tDCS therapy may improve ACh’s concentration and exerts benefits in AD. Furthermore, this effect even lasts 2 months after tDCS [243]. In addition to its impact on ChAT and ACh, tDCS can also affect other neurotransmitters [271]. In healthy individuals, the effect of tDCS is mediated by multiple monoamine neurotransmitters (e.g., dopamine and serotonin) [272,273,274], and the regulation of these neurotransmitters may contribute to the neuroprotective role of tDCS in AD.

Ameliorates neuroinflammation

tDCS also displays pronounced anti-inflammatory effects in AD. In AD, the deposition of Aβ oligomers induces astrocyte activation [9, 44]. The excessive activation of astrocytes will release a large number of pro-inflammatory, which in turn facilitate the deposition of amyloid plaques [9, 44]. In an AD mouse model, tDCS reduces astrocyte activation in different brain areas, including CA1-CA3 and dentate gyrus [251]. Not only in AD, but the anti-inflammatory effects of tDCS have also been found in other dementia [275]. For example, in a rat model of vascular dementia, anodal tDCS significantly suppressed the over-activation of microglia and astrocyte, accompanied by attenuation of demyelination in both the corpus callosum and internal capsule [275]. Consistent with the changes in glial cells, the increased levels of pro-inflammatory factors (e.g., IL-1β, IL-6, and TNF-α) were alleviated by anodal tDCS [275]. Interestingly, cathodal and anodal tDCS promote microglial activation in the healthy brain [276]. However, in an ischemic stroke, the effect of tDCS on microglia is different from that in the healthy brain [277]. A study showed that cathodal tDCS could reduce the excessive activation of microglial and alleviate inflammation in the peri-ischemic cortex [277]. Therefore, the effects of tDCS may also depend on the health conditions of the brain.

Challenges of tDCS therapy in AD

Similar to PBM therapy and rTMS, the effects of tDCS depend on the intensity, the duration, the position of the electrode over the scalp, the number of sessions of tDCS, and the mode of stimulation [235]. Although a more significant number of clinical trials have been performed to measure the effects of tDCS on AD, there are no standard protocols regarding the clinical use of tDCS [278].

Another question is how to extend the after-effect. As mentioned previously, the after-effect of the tDCS usually only lasts several minutes [247]. Studies that focus on finding the optimal parameters to extend the after-effect of the tDCS may help improve the beneficial effect of tDCS.

Third, the effects of cathodal and anodal tDCS in the different animal models are inconsistent [275, 277]. Although, as mentioned previously, most studies only found beneficial effects of anodal tDCS [245, 246]. Inconsistences still exist. For example, a previous study found that cathodal stimulation could protect cortical neurons against ischemic damage and facilitate clinical recovery [277]. However, anodal tDCS deteriorated postischemic lesion volume and augmented the derangement of the blood-brain barrier [277]. Therefore, more studies are still needed to clarify the effect of anodal tDCS and cathodal tDCS.

Finally, more studies are required to investigate the specific target of tDCS. For example, there is a very close association between light, electricity, and magnetism [279]. Mitochondria is a primary target of PBM and a mediator of rTMS, but studies investigating the effect of tDCS on mitochondria in AD are less numerous. However, other neurodegenerative diseases provided insights into the mitochondrial changes with tDCS therapy in AD [280]. For example, in a Parkinson’s disease (PD) mouse model, the PD animal showed pronounced mitochondrial damage, as evidenced by mitochondrial swelling, decreased mitochondrial glutamate dehydrogenase, and decreased ATP content in the substantianigra [280]. In addition, the PD mouse model also displayed reduced mitochondrial biogenesis-related protein and significantly elevated fission-associated protein (Drp1), indicating mitochondrial biogenesis deficits and excessive mitochondrial fragmentation [280]. Interestingly, these changes were alleviated by anodal tDCS, which implies a potential role of tDCS in protecting mitochondria [280]. However, to the best of our knowledge, we did not find research investigating whether mitochondria are one of the potential targets of tDCS.

Exercise and parameters of intervention

Substantial evidence indicates that physical excise is a known modifiable risk factor for AD [281]. In addition, physical exercise slows down AD progression and development and helps combat other AD risk factors and complications [9, 44, 282]. Many studies demonstrate the neuroprotective of exercise interventions [9, 44, 281, 282]. Compared with other pharmacological and other non-invasive therapies, physical exercise is easily accessible and affordable [281]. The effects of exercise can be affected by four elements, including intensity, time, frequency, and type of exercise [283]. Physical activity can be categorized into low, moderate, and vigorous intensity measured according to the heart rate or percentage of the maximal oxygen consumption (can be measured by the metabolic equivalent of task) [284]. In AD, the type of exercise intervention is also an essential factor affecting exercise benefits [285]. The types of exercise can currently be classified into four categories: strength, flexibility, endurance, and balance. In AD studies, aerobic exercise is the most well-studied endurance exercise and displays beneficial effects [9, 44]. According to previous studies, exercise interventions can improve cognitive function, improve neuroplasticity, and reduce neuropsychiatric symptoms [9, 44]. Similar to other non-invasive theories, exercise also benefits AD treatment or prevention [9, 44].

Exercise therapy for AD

Alleviates learning and memory deficits and anxious-depressive-like behaviors

A large body of studies has provided evidence supporting the beneficial effects of physical excise in preserving learning and memory function [9, 44]. In multiple animal models of AD, physical exercise has worked as a pre-/post-treatment strategy in improving cognitive function or preventing memory loss [9, 44]. For example, in the STZ-induced sporadic AD rat model, animals for 4-week post-intervention using treadmill exercise displayed significantly improved hippocampal-dependent cognitive functioning, including the improved spatial learning and memory and recognition memory [286]. Similarly, in a transgenic AD mouse model with Tau pathology, after 9 months of exercise, AD-like mice from the long-term voluntary exercise groups showed significant improvement in short-term working memory [287]. Moreover, even shorter voluntary activity for 5 months also confers a beneficial role in improving learning and memory [287]. In APP/PS1 mouse model, 12-week treadmill exercise significantly improved cognitive function, suggesting the improvement of cognitive function with physical exercise can be found in different types of physical activity with different exercise intensity or duration [288]. In addition to exercise post-treatment, increasing evidence demonstrated the preventive effect of exercise on cognitive deficits [44]. For example, 4-week swimming exercise pretreatment significantly attenuated STZ-induced learning and memory deficits, including hippocampus-dependent spatial learning and memory and recognition memory [44]. Furthermore, consistent with the improvements in cognitive function, the expressions of synaptic proteins were well preserved in the exercise pretreatment group [44].

According to previous studies, anxious-depressive-like behavior worked as both a predictor and a causal factor contributing to AD progression [9, 289]. In clinical studies, AD patients had significantly increased anxious-depressive symptoms, and individuals with psychological distress or anxiety are more likely to be diagnosed with AD [289,290,291,292]. Moreover, anxious-depressive behaviors are closely related to extensive amyloid plaques and neurofibrillary tangles [9, 290, 293]. Interestingly, exercise pretreatment attenuated anxious-depressive-like behavior and reduced typical pathological changes at the early stages of AD [192]. However, cognitive deficits did not occur at that time, suggesting physical exercise is a practical approach to prevent early-stage behavioral changes in the transgenic AD rat model [192]. Similarly, 6 months of voluntary physical training also shows its beneficial effect in ameliorating anxiety levels in a transgenic AD mouse model. Consistent with the results of animal studies, clinical trials show that AD patients with depression displayed a clear benefit after participating in the physical exercise [294]. However, more studies with a larger sample are still needed to clarify the benefits of different types of exercise in AD [294].

Increases cerebral blood flow

According to a previous study, AD manifests as a significant decrease in cerebral blood flow (CBF) [295]. Compared to age-matched healthy controls, the total blood flow of AD patients was reduced by 40% [296]. Even patients with mild cognitive impairment (MCI) have a significant decrease in CBF [297]. In AD, several specific regions are more likely to show CBF decreases, including the hippocampus, a critical area for memory [296,297,298]. CBF is closely related to the severity of AD patients and is a possible target in healthy elderly or MCI patients [297, 299]. Previous findings have suggested the association between Aβ accumulation and CBF among elderly adults across the span from controls to AD patients [300]. The higher Aβ deposition is associated with more significant CBF reduction and worse memory [300]. The increase of cerebral blood flow during exercise has been well documented in healthy individuals [301, 302], although a few studies showed a decrease or no change in CBF during physical exercise [303, 304]. Multiple factors led to the increase of CBF during physical activity [305], including the changes in arterial partial pressures of carbon dioxide (pCO2), arterial pressure, cardiac output, muscle mechanoreceptors, oxidative metabolism, and neural innervation [305, 306]. Muscle mechanoreceptors contribute to the increase of CBF at the initial exercise stage, indicating physical exercise’s beneficial role in attenuating AD pathological changes [307]. The pCO2 regulates CBF through pH changes by modulating the cerebral vessel diameter after initiating physical activity [308]. In the human study, increased CBF was found in AD patients after 12 months of aerobic exercise training [309], suggesting that the increased CBF after exercise may contribute to the effects of physical activity on the brain.

Inhibits the release of inflammatory factors and gliosis

As mentioned previously, the increased/decreased release of pro-inflammatory/anti-inflammatory factors and the changes in glial cell phenotype are implicated in the pathogenesis and development of AD [9, 22, 310]. In the STZ-induced rat model of sporadic AD, the pro-inflammatory factors (e.g., IL-1β, TNFα) are significantly increased, and the anti-inflammatory factors (e.g., Il-4, IL-10) are decreased in the hippocampal CA1 region [286]. However, 4-week treadmill exercise post-treatment alleviated these changes and promoted microglial polarization from M1 pro-inflammatory phenotype to M2 anti-inflammatory phenotype [286]. Similarly, in the STZ-induced rat model of sporadic AD, exercise pretreatment also displayed its anti-inflammatory effects [44]. In one previous study, 4 weeks of treadmill training pretreatment significantly ameliorated reactive gliosis and neuroinflammation [44]. Although the switch between M1 and M2 phenotype was not mentioned, the increased release of anti-inflammatory cytokines and decreased pro-inflammatory cytokines were found in the exercise pretreated group [44]. In addition to regulating the microglial phenotype, physical exercise also modulates the astrocytic phenotypes [311]. In the brain of AD patients, neurotoxic phenotype A1 was predominant [61, 312]. Interestingly, exercise promotes the transformation of astrocytes from neurotoxic A1 phenotype to neuroprotective A2 phenotype [311].

In most AD animal studies, exercise suppresses the overactivation of glial cells, including astrocyte and microglia, which is consistent with inflammatory factors [9, 44]. However, the study also found increased astrocytes in the hippocampus after long-term voluntary physical exercise [313]. Interestingly, the beneficial effects of physical training on gliosis are supported by reduced A1 markers and increased A2 markers [313]. Consistent with astrocyte changes, astrocyte-derived BDNF was significantly increased, suggesting astrocyte remodeling after exercise is implicated in the neuroprotective effect of physical activity [313].

Promotes oxidative stress-related adaptations and alleviates oxidative damage

Increasing evidence strongly supports a general anti-oxidative role of exercise in AD [44]. In an aged AD-like mouse model, analysis of brain tissue showed high levels of oxidative damage [314]. The excessive produced ROS reacts with protein, lipids, and nucleic acids to generate protein carbonyls, lipid peroxides, and nucleic acid oxidative modification [44]. However, the oxidative damage was significantly ameliorated by exercise by maintaining redox homeostasis [314]. Furthermore, network analysis of oxidative stress markers and AD-like pathologies showed that oxidative stress might be a major event and a primary target for exercise neuroprotection against AD pathological changes [314]. In the STZ-induced sporadic AD animal model, 4-week treadmill exercise led to a robust inhibition of oxidative damage to protein, lipids, and nucleic acids, which contributes to the neuroprotection and cognitive improvement after exercise post-treatment [286]. Moreover, exercise pretreatment also has an anti-oxidative effect in the sporadic AD animal model [44]. Similar to exercise post-treatment, swimming exercise pretreatment significantly attenuated oxidative damage to proteins and lipids [44]. In addition, exercise preserved total anti-oxidative capacity and improved the expression of antioxidant enzymes, including superoxide dismutase 2 (SOD2), thioredoxin 2 (TRX2), heme oxygenase 1 (HO-1), and NAD(P)H dehydrogenase [quinone] (NQO1) [44]. Further analysis found increased nuclear erythroid 2-related factor 2 (Nrf2) DNA-binding activity in the exercise pretreatment group [44]. Nrf2 is a critical molecule in regulating the cellular antioxidant ability [315]. In AD, the protective role of Nrf2 is compromised [44, 315]. However, exercise pretreatment promotes the translocation of Nrf2 from the cytoplasm into the nucleus and binds to the antioxidant response element (ARE) to active expressions of antioxidant enzymes, which is consistent with the changes of SOD2, TRX2, HO-1, and NAD(P)H dehydrogenase [quinone] (NQO1) after exercise treatment [44]. The anti-oxidative effect of exercise is also found in numerous human clinical trials [316, 317]. For example, a previous study found reduced ROS levels, increased antioxidant capacity, and decreased oxidative damage by-products in the blood or urine samples after 6 weeks of physical exercise [316, 317].

Improves glymphatic clearance

The glymphatic system is a glial-dependent waste clearance system that utilizes an astroglia-formed system of the perivascular channels to remove the toxins and metabolic waste from the brain in the central nervous system (CNS) of vertebrates [318]. The brain’s glymphatic system’s disruption or compromised function is implicated in the deposition of misfolded proteins in aging and neurodegenerative disease [318]. In the glymphatic system, the paravascular cerebrospinal fluid (CSF)-interstitial fluid (ISF) exchange plays a central role in the clearance of interstitial solutes [319]. The exchange of ISF and CSF along perivascular spaces depends on Aquaporin 4 (AQP4), a protein located in the end-feet of the astrocyte, which is the major component of the glymphatic system [320]. The transgenic mice lacking AQP-4 exhibit significantly declined interstitial solute clearance compared with wild animals [319]. Under physiological conditions, the CSF flows across highly polarized AQP4 molecules at astrocyte end-feet, mixing with ISF, which allows the clearance of Aβ [321, 322]. However, under pathological conditions in AD, both the AQP4 expression and polarization are reduced, and AQP4 is redistributed from the end-feet of the astrocyte to the cell body, which induces impaired ISF drainage and compromised glymphatic clearance [321,322,323]. In the APP/PS1 AD mouse model, AQP4 knockout exacerbated AD mice’s cognitive deficits and other typical pathological changes [324]. Interestingly, in aged mice with cognitive deficits, 6-week voluntary exercise promotes glymphatic clearance of Aβ by regulating AQP4 polarization and improving the expression of AQP4 in the perivascular regions [322]. In addition, the increased heart rate, perfusion flow, cerebral pulse pressure, and brain interstitial fluid flow after exercise also suggest that the improvement of glymphatic clearance mediates the neuroprotective effects of exercise [319, 325]. Moreover, although a previous study found that voluntary wheel running could not affect BBB permeation, the glymphatic clearance of Aβ and the BBB disruption have been improved following physical exercise, indicating exercise may promote BBB-mediated clearance of Aβ or tau [322].

Preserves mitochondrial bioenergetics, mitochondrial biogenesis, and quality control

Mitochondrial adaptations are another crucial underlying mechanism for exercise-induced neuroprotection [326]. Exercise-induced mitochondrial adaptations include preserved mitochondrial bioenergetics, mitochondrial biogenesis, and mitochondrial quality control [326]. The decreased activity of enzymes in the mitochondrial electron transport chain (ETC), dysregulation of calcium homeostasis, reduced antioxidant enzymatic activity, and increased ROS generation led to mitochondria dysfunction and, therefore, decreased ATP synthesis [22, 44, 327]. However, in the brain of aged mice, a total of 17 days of treadmill training significantly improved the activity of enzymes in ETC and increased rates of mitochondrial respiration and ROS production [328]. Notably, the increased ROS generation after exercise differs from the pathological ROS accumulation [329]. Exercise-induced ROS generation is a moderate and short-term increase, which induces mitochondrial adaptions to oxidative stress and improves the brain’s enzymatic antioxidant system [329]. Intriguingly, in the transgenic mouse AD model, the pathological increased hippocampal ROS accumulation in the AD mouse model was reduced after long-term treadmill running [330]. In the same AD animal model, the impaired mitochondrial function was restored by 20-week treadmill running, as evidenced by increased mitochondrial state 3 and 4 respiration [330]. Furthermore, the impaired complexes I, IV, and ATP synthase activities were improved in the hippocampus of AD mice with exercise training [330]. This evidence demonstrates that exercise training preserves mitochondrial bioenergetics and exerts neuroprotection in AD [328, 330].

Mitochondrial biogenesis is the guarantee for the number of mitochondria and the energy requirement in neurons [331]. However, mitochondrial biogenesis is impaired in AD by increased mtDNA mutations and decreased peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) [332]. In addition, mitochondrial biogenesis requires the coordinated synthesis of proteins encoded by mtDNA and nuclear DNA [331]. However, the increased mtDNA mutations and decreased expression of PGC-1α and its downstream target have been widely reported in AD [22, 331]. Intriguingly, 6-month exercise training alleviates the damaged mtDNA in the transgenic AD mouse model [333]. Similarly, in the APP/PS1 transgenic mouse model, 20-week treadmill training improves mtDNA repairability in the mouse hippocampus, which protects against the impairment of mitochondrial function and mitochondrial biogenesis [330].

As mentioned previously, the balance between mitochondrial fission and fusion is crucial for maintaining a healthy mitochondrial pool and is essential for cellular function and stress responses [28]. AD disrupts this balance, causing excessive mitochondrial fragmentation and impaired mitochondrial function [22]. However, evidence suggests that exercise regulates mitochondrial dynamics and maintains mitochondrial homeostasis [331, 334]. A previous study found that under physiological conditions, the phosphorylation of mitochondrial fission-related protein Drp1 was increased immediately after acute resistance exercise, suggesting the acute exercise triggers the self-protective mechanism against mitochondrial damage by segregating the damaged mitochondria [335]. In contrast, long-term resistance training improves mitochondrial fusion-related protein to prevent excessive mitochondrial fission and maintain mitochondrial function [335]. In APP/PS1 transgenic mice, moderate-intensity continuous training and high-intensity interval training down-regulated the expression of the mitochondrial fission-related protein (e.g., Drp1 and Fis1). In contrast, the expression of mitochondrial fusion-related proteins (e.g., MFN1, MFN2, and OPA1) was improved, accompanied by alleviation of cognitive dysfunction, enhanced the activity of antioxidant enzymes, and decreased oxidative stress [336].

Interestingly, improved mitochondrial quality control after exercise training was found in other aging-related diseases, including sporadic inclusion body myositis and Parkinson’s disease [337, 338]. Furthermore, in a human study, both the mitochondrial fission and fusion-related proteins were increased in the active subjects compared with the respective sedentary ones, suggesting exercise promotes the efficiency of mitochondrial quality control by improving the expressions of mitochondrial dynamics associated proteins [339]. Moreover, research on an AD mouse model found that the efficiency of exercise therapy on mitochondrial dynamics was regulated by PGC1a/FNDC5/irisin pathway, a sensitive pathway in response to physical exercise, suggesting the potential role of exercise on mitochondrial dynamics in AD [340].

Challenges of exercise therapy in AD

Similar to other non-invasive approaches, questions still focused on which intensity is the optimal dosage? Which is the best exercise form for preventing or slowing down AD? Furthermore, another question that remains unanswered is investigating the optimal timing of initiation of physical exercise treatment. The earlier the exercise treatment begins, the better the outcomes, or does the individual enjoy a similar beneficial effect even to exercise after mid-life or later life? Additionally, although numerous studies support the beneficial role of exercise in AD, a study found that exercise appears not to affect working memory deficit [341]. Therefore, more studies are still needed to clarify the effects of physical activity on different types of memory tasks. To date, although significant progress has been achieved in animal studies, no randomized controlled trials demonstrate that exercise can prevent dementia [9, 44, 282, 342]. For example, according to a previous study, there are no noticeable improvements in cognitive function in people with mild to moderate dementia [342]. Therefore, more studies on humans are needed to determine why the effects of exercise are so variable. Additionally, a previous study found that 6 months of exercise intervention had gender differences in cognition. The women performed better on cognitive function-based tasks than men [343]. Therefore, gender difference is a major factor that must be considered in AD studies. Finally, more animal and human studies are still required to elucidate the underlying molecular mechanism of physical activity in AD [310]. For instance, the BBB breakdown has been confirmed in both the AD animal and postmortem AD patients [344]. However, to the best of our knowledge, we did not find enough evidence to demonstrate the effectiveness of exercise on BBB.

Summary of effects, mechanism, and future directions

As discussed previously, these non-invasive approaches are promising therapeutic methods for slowing AD progression or alleviating AD typical symptoms [9, 44, 69, 70, 240, 243]. For example, the studies discussed offer compelling evidence that non-invasive approaches, including PBM, rTMS, tDCS, and physical exercise, can alleviate memory loss and reduce pathological hallmarks of AD (e.g., amyloid plaques and tau pathology) [69, 102, 251]. In addition, as shown in Fig. 7, the beneficial effects of these non-invasive treatments include alleviating neuronal apoptosis, improving cerebral perfusion, recruiting microglia around amyloid plaques, improving microglial phagocytosis, attenuating oxidative stress, regulating LTP and LTD, promoting neurogenesis, preserving synaptic proteins, alleviating mtDNA damage, improving glymphatic clearance, regulating neurotransmitters, increasing neurotrophins, maintaining mitochondrial function and mitochondrial dynamics, promoting the transformation of glial cells from neurotoxic to neuroprotective phenotype, and enhancing angiogenesis. Although these factors that contribute to the beneficial effects of non-invasive treatments seem independent, they are interconnected and affect each other. For instance, the improvement of the microenvironment significantly enhances neurogenesis, promotes angiogenesis, and alleviates neuronal apoptosis [10, 121]. Mitochondrial function, mitochondria-associated oxidative stress, and neuroinflammation are central factors in modulating the neuronal microenvironment [22].

Fig. 7
figure 7

Effects and mechanisms for non-invasive therapies in AD. These non-invasive treatments are able to alleviate neuronal apoptosis, improve cerebral perfusion, recruit microglia around amyloid plaques, improve microglial phagocytosis, attenuate oxidative stress, regulate LTP and LTD, promote neurogenesis, preserve synaptic proteins, alleviate mtDNA damage, improve glymphatic clearance, regulate neurotransmitters, increase neurotrophins, maintain mitochondrial function and mitochondrial dynamics, promote the transformation of glial cells from neurotoxic to neuroprotective phenotype, and promote angiogenesis

The failure of pharmacological therapy targeting only single AD-related pathology suggests that multi-target therapy is a potential approach to AD treatment [345]. Non-invasive therapies discussed in this review target multiple pathological changes in AD and display promising effects in improving cognitive function and anxious-depressive-like behavior. Therefore, to improve the translational value of these non-invasive therapies, it will be critical to understand the underlying mechanisms and find the optimal therapy parameters in clinical application. Moreover, the combined use of non-invasive therapies is also a promising research direction for future studies [346].

Conclusions

Non-invasive therapies have the potential and promise to achieve therapeutic goals of slowing down or preventing AD. As shown in Table 1, the improvements in cognitive function and other behavioral changes after non-invasive treatments rely on multiple cellular changes and adaptive responses. The challenges to advancing non-invasive treatments in AD prevention and treatment are shown in Table 2. No agreement on the optimal therapy parameters in clinical application and finding the exact or specific targets are the primary challenges for future studies. If future studies are successful in these non-invasive approaches, they will shed light on AD treatment and prevention.

Table 1 Effects of non-invasive therapy for Alzheimer’s disease
Table 2 Challenges for non-invasive therapies in AD treatment

Availability of data and materials

Not applicable.

Abbreviations

AD:

Alzheimer’s disease

PBM:

Photobiomodulation

TMS:

Transcranial magnetic stimulation

tDCS:

Transcranial direct current stimulation

APP:

Amyloid precursor protein

NFTs:

Neurofibrillary tangles

ATP:

Adenosine triphosphate

ROS:

Reactive oxygen species

DAMPs:

Damage-associated molecular patterns

IL-3:

Interleukin-3

PS1:

Presenilin-1

LLLT:

Low-level laser (light) therapy

CCO:

Cytochrome C oxidase

PHF1:

Hyperphosphorylated tau

NIR:

Near-infrared light

NO:

Nitric oxide

4HNE:

4-Hydroxyhexenal

8-OHDG:

8-Hydroxydeoxyguanosine

rTMS:

Repetitive transcranial magnetic stimulation

MDD:

Major depressive disorder

FDA:

Food and Drug Administration

TBS:

Theta-burst stimulation

MCI:

Mild cognitive impairment

LTP:

Long-term potentiation

LTD:

Long-term depression

BDNF:

Brain-derived neurotrophic factor

TrkB:

Tropomyosin-related kinase B

5-HT:

Serotonin

DLPFC:

Dorsolateral prefrontal cortex

GABA:

Gamma-aminobutyric acid

OGD:

Oxygen-glucose deprivation

ChAT:

Choline acetyltransferase

Ach:

Acetylcholine

PD:

Parkinson’s disease

CBF:

Cerebral blood flow

SOD2:

Superoxide dismutase 2

TRX2:

Thioredoxin 2 (TRX2)

HO-1:

Heme oxygenase 1 (HO-1)

NQO1:

NAD(P) H dehydrogenase [quinone]

Nrf2:

Nuclear erythroid 2-related factor 2

ARE:

Antioxidant response element

CNS:

Central nervous system

AQP4:

Aquaporin 4

ETC:

Electron transport chain

PGC-1α:

Proliferator-activated receptor gamma coactivator 1-alpha

BBB:

Blood-brain barrier

References

  1. Liu PP, Xie Y, Meng XY, Kang JS. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct Target Ther. 2019;4:29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14:32.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Association Alzheimer’s. Alzheimer’s disease facts and figures. Alzheimers Dement. 2021;17:327–406.

    CAS  Google Scholar 

  4. Haque RU, Levey AI. Alzheimer’s disease: a clinical perspective and future nonhuman primate research opportunities. Proc Natl Acad Sci U S A. 2019;116(52):26224–9.

    Article  CAS  PubMed Central  Google Scholar 

  5. Pandey G, Ramakrishnan V. Invasive and non-invasive therapies for Alzheimer’s disease and other amyloidosis. Biophys Rev. 2020;12:1175–86.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Dompe C, Moncrieff L, Matys J, Grzech-Lesniak K, Kocherova I, Bryja A, et al. Photobiomodulation-underlying mechanism and clinical applications. J Clin Med. 2020;9:1724.

    Article  PubMed Central  Google Scholar 

  7. Yang L, Wu C, Tucker L, Dong Y, Li Y, Xu P, et al. Photobiomodulation therapy attenuates anxious-depressive-like behavior in the TgF344 rat model. J Alzheimers Dis. 2021;83:1415–29.

    Article  CAS  PubMed  Google Scholar 

  8. Yang L, Dong Y, Wu C, Youngblood H, Li Y, Zong X, et al. Effects of prenatal photobiomodulation treatment on neonatal hypoxic ischemia in rat offspring. Theranostics. 2021;11:1269–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wu C, Yang L, Li Y, Dong Y, Yang B, Tucker LD, et al. Effects of exercise training on anxious-depressive-like behavior in Alzheimer rat. Med Sci Sports Exerc. 2020;52:1456–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zong X, Dong Y, Li Y, Yang L, Li Y, Yang B, et al. Beneficial effects of theta-burst transcranial magnetic stimulation on stroke injury via improving neuronal microenvironment and mitochondrial integrity. Transl Stroke Res. 2020;11:450–67.

    Article  CAS  PubMed  Google Scholar 

  11. Chang CH, Lane HY, Lin CH. Brain stimulation in Alzheimer’s disease. Front Psychiatry. 2018;9:201.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kametani F, Hasegawa M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front Neurosci. 2018;12:25.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zheng H, Koo EH. Biology and pathophysiology of the amyloid precursor protein. Mol Neurodegener. 2011;6:27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol Sci. 1991;12:383–8.

    Article  CAS  PubMed  Google Scholar 

  15. Nunan J, Small DH. Regulation of APP cleavage by alpha-, beta- and gamma-secretases. FEBS Lett. 2000;483:6–10.

    Article  CAS  PubMed  Google Scholar 

  16. Chow VW, Mattson MP, Wong PC, Gleichmann M. An overview of APP processing enzymes and products. Neuromolecular Med. 2010;12:1–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Miao J, Shi R, Li L, Chen F, Zhou Y, Tung YC, et al. Pathological tau from Alzheimer’s brain induces site-specific hyperphosphorylation and SDS- and reducing agent-resistant aggregation of tau in vivo. Front Aging Neurosci. 2019;11:34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gong CX, Iqbal K. Hyperphosphorylation of microtubule-associated protein tau: a promising therapeutic target for Alzheimer disease. Curr Med Chem. 2008;15:2321–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Barbier P, Zejneli O, Martinho M, Lasorsa A, Belle V, Smet-Nocca C, et al. Role of tau as a microtubule-associated protein: structural and functional aspects. Front Aging Neurosci. 2019;11:204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Alonso AD, Cohen LS, Corbo C, Morozova V, ElIdrissi A, Phillips G, et al. Hyperphosphorylation of tau associates with changes in its function beyond microtubule stability. Front Cell Neurosci. 2018;12:338.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yuan A, Sershen H, Veeranna BBS, Kumar A, Hashim A, et al. Neurofilament subunits are integral components of synapses and modulate neurotransmission and behavior in vivo. Mol Psychiatry. 2015;20:986–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang L, Youngblood H, Wu C, Zhang Q. Mitochondria as a target for neuroprotection: role of methylene blue and photobiomodulation. Transl Neurodegener. 2020;9:19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener. 2020;15:30.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Tucker LD, Lu Y, Dong Y, Yang L, Li Y, Zhao N, et al. Photobiomodulation therapy attenuates hypoxic-ischemic injury in a neonatal rat model. J Mol Neurosci. 2018;65:514–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Youle RJ, van der Bliek AM. Mitochondrial fission, fusion, and stress. Science. 2012;337:1062–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Fu W, Liu Y, Yin H. Mitochondrial dynamics: biogenesis, fission, fusion, and mitophagy in the regulation of stem cell behaviors. Stem Cells Int. 2019;2019:9757201.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sun X, Alford J, Qiu H. Structural and functional remodeling of mitochondria in cardiac diseases. Int J Mol Sci. 2021;22:4167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ren L, Chen X, Chen X, Li J, Cheng B, Xia J. Mitochondrial dynamics: fission and fusion in fate determination of mesenchymal stem cells. Front Cell Dev Biol. 2020;8:580070.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Chen H, Chan DC. Emerging functions of mammalian mitochondrial fusion and fission. Hum Mol Genet. 2005;14 Spec No. 2:R283–9.

    Article  PubMed  Google Scholar 

  30. Wang W, Yin J, Ma X, Zhao F, Siedlak SL, Wang Z, et al. Inhibition of mitochondrial fragmentation protects against Alzheimer’s disease in rodent model. Hum Mol Genet. 2017;26:4118–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Manczak M, Calkins MJ, Reddy PH. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: implications for neuronal damage. Hum Mol Genet. 2011;20:2495–509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD. Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106:14670–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gibson GE, Sheu KF, Blass JP. Abnormalities of mitochondrial enzymes in Alzheimer disease. J Neural Transm (Vienna). 1998;105:855–70.

    Article  CAS  PubMed  Google Scholar 

  34. Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G. Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta. 2010;1802:2–10.

    Article  CAS  PubMed  Google Scholar 

  35. Swerdlow RH. Mitochondria and mitochondrial cascades in Alzheimer’s disease. J Alzheimers Dis. 2018;62:1403–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Armand-Ugon M, Ansoleaga B, Berjaoui S, Ferrer I. Reduced mitochondrial activity is early and steady in the entorhinal cortex but it is mainly unmodified in the frontal cortex in Alzheimer’s disease. Curr Alzheimer Res. 2017;14:1327–34.

    Article  CAS  PubMed  Google Scholar 

  37. Huang WJ, Zhang X, Chen WW. Role of oxidative stress in Alzheimer’s disease. Biomed Rep. 2016;4:519–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao RZ, Jiang S, Zhang L, Yu ZB. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med. 2019;44:3–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Brand MD. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic Biol Med. 2016;100:14–31.

    Article  CAS  PubMed  Google Scholar 

  40. D'Autreaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007;8:813–24.

    Article  CAS  PubMed  Google Scholar 

  41. Teixeira JP, de Castro AA, Soares FV, da Cunha EFF, Ramalho TC. Future therapeutic perspectives into the Alzheimer’s disease targeting the oxidative stress hypothesis. Molecules. 2019;24:4410.

    Article  CAS  PubMed Central  Google Scholar 

  42. Srinivasan S, Avadhani NG. Cytochrome c oxidase dysfunction in oxidative stress. Free Radic Biol Med. 2012;53:1252–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pao PC, Patnaik D, Watson LA, Gao F, Pan L, Wang J, et al. HDAC1 modulates OGG1-initiated oxidative DNA damage repair in the aging brain and Alzheimer’s disease. Nat Commun. 2020;11:2484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wu C, Yang L, Tucker D, Dong Y, Zhu L, Duan R, et al. Beneficial effects of exercise pretreatment in a sporadic Alzheimer’s rat model. Med Sci Sports Exerc. 2018;50:945–56.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Chen Z, Zhong C. Oxidative stress in Alzheimer’s disease. Neurosci Bull. 2014;30:271–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hensley K. Neuroinflammation in Alzheimer’s disease: mechanisms, pathologic consequences, and potential for therapeutic manipulation. J Alzheimers Dis. 2010;21:1–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lai AY, McLaurin J. Clearance of amyloid-beta peptides by microglia and macrophages: the issue of what, when and where. Future Neurol. 2012;7:165–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Fakhoury M. Microglia and astrocytes in Alzheimer’s disease: implications for therapy. Curr Neuropharmacol. 2018;16:508–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ismail R, Parbo P, Madsen LS, Hansen AK, Hansen KV, Schaldemose JL, et al. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: a longitudinal PET study. J Neuroinflammation. 2020;17:151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2016;53:1181–94.

    Article  CAS  PubMed  Google Scholar 

  51. Shen Z, Bao X, Wang R. Clinical PET imaging of microglial activation: implications for microglial therapeutics in Alzheimer’s disease. Front Aging Neurosci. 2018;10:314.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mandrekar-Colucci S, Landreth GE. Microglia and inflammation in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010;9:156–67.

    Article  CAS  PubMed  Google Scholar 

  53. Malm TM, Jay TR, Landreth GE. The evolving biology of microglia in Alzheimer’s disease. Neurotherapeutics. 2015;12:81–93.

    Article  CAS  PubMed  Google Scholar 

  54. Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms underlying the rapid peroxisome proliferator-activated receptor-gamma-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer’s disease. J Neurosci. 2012;32:10117–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hsiao CC, Sankowski R, Prinz M, Smolders J, Huitinga I, Hamann J. GPCRomics of homeostatic and disease-associated human microglia. Front Immunol. 2021;12:674189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169:1276–90 e1217.

    Article  CAS  PubMed  Google Scholar 

  57. Frost GR, Li YM. The role of astrocytes in amyloid production and Alzheimer’s disease. Open Biol. 2017;7:170228.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Kashon ML, Ross GW, O'Callaghan JP, Miller DB, Petrovitch H, Burchfiel CM, et al. Associations of cortical astrogliosis with cognitive performance and dementia status. J Alzheimers Dis. 2004;6:595–604 discussion 673-581.

    Article  PubMed  Google Scholar 

  59. Mrak RE, Sheng JG, Griffin WS. Correlation of astrocytic S100 beta expression with dystrophic neurites in amyloid plaques of Alzheimer’s disease. J Neuropathol Exp Neurol. 1996;55:273–9.

    Article  CAS  PubMed  Google Scholar 

  60. Beach TG, McGeer EG. Lamina-specific arrangement of astrocytic gliosis and senile plaques in Alzheimer’s disease visual cortex. Brain Res. 1988;463:357–61.

    Article  CAS  PubMed  Google Scholar 

  61. Gonzalez-Reyes RE, Nava-Mesa MO, Vargas-Sanchez K, Ariza-Salamanca D, Mora-Munoz L. Involvement of astrocytes in Alzheimer’s disease from a neuroinflammatory and oxidative stress perspective. Front Mol Neurosci. 2017;10:427.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Grimaldi A, Pediconi N, Oieni F, Pizzarelli R, Rosito M, Giubettini M, et al. Neuroinflammatory processes, A1 astrocyte activation and protein aggregation in the retina of Alzheimer’s disease patients, possible biomarkers for early diagnosis. Front Neurosci. 2019;13:925.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Joshi AU, Minhas PS, Liddelow SA, Haileselassie B, Andreasson KI, Dorn GW 2nd, et al. Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat Neurosci. 2019;22:1635–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Neal M, Luo J, Harischandra DS, Gordon R, Sarkar S, Jin H, et al. Prokineticin-2 promotes chemotaxis and alternative A2 reactivity of astrocytes. Glia. 2018;66:2137–57.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Reichenbach N, Delekate A, Plescher M, Schmitt F, Krauss S, Blank N, et al. Inhibition of Stat3-mediated astrogliosis ameliorates pathology in an Alzheimer’s disease model. EMBO Mol Med. 2019;11:e9665.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Ceyzeriat K, Ben Haim L, Denizot A, Pommier D, Matos M, Guillemaud O, et al. Modulation of astrocyte reactivity improves functional deficits in mouse models of Alzheimer’s disease. Acta Neuropathol Commun. 2018;6:104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Daria A, Colombo A, Llovera G, Hampel H, Willem M, Liesz A, et al. Young microglia restore amyloid plaque clearance of aged microglia. EMBO J. 2017;36:583–603.

    Article  CAS  PubMed  Google Scholar 

  69. Tao L, Liu Q, Zhang F, Fu Y, Zhu X, Weng X, et al. Microglia modulation with 1070-nm light attenuates Abeta burden and cognitive impairment in Alzheimer’s disease mouse model. Light Sci Appl. 2021;10:179.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, et al. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature. 2016;540:230–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. McAlpine CS, Park J, Griciuc A, Kim E, Choi SH, Iwamoto Y, et al. Astrocytic interleukin-3 programs microglia and limits Alzheimer’s disease. Nature. 2021;595:701–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Dorostkar MM, Burgold S, Filser S, Barghorn S, Schmidt B, Anumala UR, et al. Immunotherapy alleviates amyloid-associated synaptic pathology in an Alzheimer’s disease mouse model. Brain. 2014;137:3319–26.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Cummings J, Ritter A, Zhong K. Clinical trials for disease-modifying therapies in Alzheimer’s disease: a primer, lessons learned, and a blueprint for the future. J Alzheimers Dis. 2018;64:S3–S22.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Frozza RL, Lourenco MV, De Felice FG. Challenges for Alzheimer’s disease therapy: insights from novel mechanisms beyond memory defects. Front Neurosci. 2018;12:37.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Chu LW. Alzheimer’s disease: early diagnosis and treatment. Hong Kong Med J. 2012;18:228–37.

    CAS  PubMed  Google Scholar 

  76. Tripathi A, Bankaitis VA. Molecular docking: from lock and key to combination lock. J Mol Med Clin Appl. 2017;2. https://doi.org/10.16966/2575-0305.106.

  77. Chen Y, Liang Z, Blanchard J, Dai CL, Sun S, Lee MH, et al. A non-transgenic mouse model (icv-STZ mouse) of Alzheimer’s disease: similarities to and differences from the transgenic model (3xTg-AD mouse). Mol Neurobiol. 2013;47:711–25.

    Article  CAS  PubMed  Google Scholar 

  78. Drummond E, Wisniewski T. Alzheimer’s disease: experimental models and reality. Acta Neuropathol. 2017;133:155–75.

    Article  CAS  PubMed  Google Scholar 

  79. Cohen RM, Rezai-Zadeh K, Weitz TM, Rentsendorj A, Gate D, Spivak I, et al. A transgenic Alzheimer rat with plaques, tau pathology, behavioral impairment, oligomeric abeta, and frank neuronal loss. J Neurosci. 2013;33:6245–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Hamblin MR. Photobiomodulation for Alzheimer’s disease: has the light dawned? Photonics. 2019;6:77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Hamblin MR. Photobiomodulation for the management of alopecia: mechanisms of action, patient selection and perspectives. Clin Cosmet Investig Dermatol. 2019;12:669–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Mester E, Szende B, Spiry T, Scher A. Stimulation of wound healing by laser rays. Acta Chir Acad Sci Hung. 1972;13:315–24.

    CAS  PubMed  Google Scholar 

  83. Hamblin MR. Photobiomodulation or low-level laser therapy. J Biophotonics. 2016;9:1122–4.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Hopkins JT, McLoda TA, Seegmiller JG, David BG. Low-level laser therapy facilitates superficial wound healing in humans: a triple-blind, sham-controlled study. J Athl Train. 2004;39:223–9.

    PubMed  PubMed Central  Google Scholar 

  85. Mester E, Mester AF, Mester A. The biomedical effects of laser application. Lasers Surg Med. 1985;5:31–9.

    Article  CAS  PubMed  Google Scholar 

  86. Mester E, Spiry T, Szende B, Tota JG. Effect of laser rays on wound healing. Am J Surg. 1971;122:532–5.

    Article  CAS  PubMed  Google Scholar 

  87. Dominguez Camacho A, Bravo Reyes M, Velasquez Cujar SA. A systematic review of the effective laser wavelength range in delivering photobiomodulation for pain relief in active orthodontic treatment. Int Orthod. 2020;18:684–95.

    Article  PubMed  Google Scholar 

  88. Langella LG, Casalechi HL, Tomazoni SS, Johnson DS, Albertini R, Pallotta RC, et al. Photobiomodulation therapy (PBMT) on acute pain and inflammation in patients who underwent total hip arthroplasty-a randomized, triple-blind, placebo-controlled clinical trial. Lasers Med Sci. 2018;33:1933–40.

    Article  PubMed  Google Scholar 

  89. Janzadeh A, Nasirinezhad F, Masoumipoor M, Jameie SB, Hayat P. Photobiomodulation therapy reduces apoptotic factors and increases glutathione levels in a neuropathic pain model. Lasers Med Sci. 2016;31:1863–9.

    Article  PubMed  Google Scholar 

  90. Leal-Junior EC. Photobiomodulation therapy in skeletal muscle: from exercise performance to muscular dystrophies. Photomed Laser Surg. 2015;33:53–4.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Li Y, Dong Y, Yang L, Tucker L, Zong X, Brann D, et al. Photobiomodulation prevents PTSD-like memory impairments in rats. Mol Psychiatry. 2021;26:6666–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hamblin MR. Shining light on the head: photobiomodulation for brain disorders. BBA Clin. 2016;6:113–24.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Wang Y, Huang YY, Wang Y, Lyu P, Hamblin MR. Red (660 nm) or near-infrared (810 nm) photobiomodulation stimulates, while blue (415 nm), green (540 nm) light inhibits proliferation in human adipose-derived stem cells. Sci Rep. 2017;7:7781.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Wang Y, Huang YY, Wang Y, Lyu P, Hamblin MR. Photobiomodulation (blue and green light) encourages osteoblastic-differentiation of human adipose-derived stem cells: role of intracellular calcium and light-gated ion channels. Sci Rep. 2016;6:33719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Ezzati K, Laakso EL, Saberi A, Yousefzadeh Chabok S, Nasiri E, Bakhshayesh EB. A comparative study of the dose-dependent effects of low level and high intensity photobiomodulation (laser) therapy on pain and electrophysiological parameters in patients with carpal tunnel syndrome. Eur J Phys Rehabil Med. 2020;56:733–40.

    PubMed  Google Scholar 

  96. Zein R, Selting W, Hamblin MR. Review of light parameters and photobiomodulation efficacy: dive into complexity. J Biomed Opt. 2018;23:1–17.

    Article  PubMed  Google Scholar 

  97. Chen Y, De Taboada L, O'Connor M, Delapp S, Zivin JA. Thermal effects of transcranial near-infrared laser irradiation on rabbit cortex. Neurosci Lett. 2013;553:99–103.

    Article  CAS  PubMed  Google Scholar 

  98. Tuner J, Jenkins PA. Parameter reproducibility in photobiomodulation. Photomed Laser Surg. 2016;34:91–2.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Carvalho AR, Caovilla AR, Ficagna A, Vieira DD, Borrero A, Bertolini GRF. Using a single dose of photobiomodulation (laser + LED) to improve performance of lower limbs in functional test: Randomized clinical trial. J Bodyw Mov Ther. 2021;28:255–63.

    Article  PubMed  Google Scholar 

  100. Liu XG, Zhou YJ, Liu TC, Yuan JQ. Effects of low-level laser irradiation on rat skeletal muscle injury after eccentric exercise. Photomed Laser Surg. 2009;27:863–9.

    Article  CAS  PubMed  Google Scholar 

  101. Keshri GK, Gupta A, Yadav A, Sharma SK, Singh SB. Photobiomodulation with pulsed and continuous wave near-infrared laser (810 nm, Al-Ga-As) augments dermal wound healing in immunosuppressed rats. PLoS One. 2016;11:e0166705.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Lu Y, Wang R, Dong Y, Tucker D, Zhao N, Ahmed ME, et al. Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiol Aging. 2017;49:165–82.

    Article  CAS  PubMed  Google Scholar 

  103. Kim HB, Baik KY, Choung PH, Chung JH. Pulse frequency dependency of photobiomodulation on the bioenergetic functions of human dental pulp stem cells. Sci Rep. 2017;7:15927.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Maksimovich IV. Laser technologies as a new direction in transcatheter interventions. Photobiomodul Photomed Laser Surg. 2019;37:455–6.

    Article  PubMed  Google Scholar 

  105. Chao LL. Effects of home photobiomodulation treatments on cognitive and behavioral function, cerebral perfusion, and resting-state functional connectivity in patients with dementia: a pilot trial. Photobiomodul Photomed Laser Surg. 2019;37:133–41.

    Article  PubMed  Google Scholar 

  106. Berman MH, Halper JP, Nichols TW, Jarrett H, Lundy A, Huang JH. Photobiomodulation with near infrared light helmet in a pilot, placebo controlled clinical trial in dementia patients testing memory and cognition. J Neurol Neurosci. 2017;8:176.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Saltmarche AE, Naeser MA, Ho KF, Hamblin MR, Lim L. Significant improvement in cognition in mild to moderately severe dementia cases treated with transcranial plus intranasal photobiomodulation: case series report. Photomed Laser Surg. 2017;35:432–41.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Peck KJ, Girard TA, Russo FA, Fiocco AJ. Music and memory in Alzheimer’s disease and the potential underlying mechanisms. J Alzheimers Dis. 2016;51:949–59.

    Article  PubMed  Google Scholar 

  109. Jahn H. Memory loss in Alzheimer’s disease. Dialogues Clin Neurosci. 2013;15:445–54.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Kelley BJ, Petersen RC. Alzheimer’s disease and mild cognitive impairment. Neurol Clin. 2007;25:577–609 v.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Salehpour F, Khademi M, Bragin DE, DiDuro JO. Photobiomodulation therapy and the glymphatic system: promising applications for augmenting the brain lymphatic drainage system. Int J Mol Sci. 2022;23:2975.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Adaikkan C, Middleton SJ, Marco A, Pao PC, Mathys H, Kim DN, et al. Gamma entrainment binds higher-order brain regions and offers neuroprotection. Neuron. 2019;102:929–43 e928.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Meng C, He Z, Xing D. Low-level laser therapy rescues dendrite atrophy via upregulating BDNF expression: implications for Alzheimer’s disease. J Neurosci. 2013;33:13505–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Dafsari FS, Jessen F. Depression-an underrecognized target for prevention of dementia in Alzheimer’s disease. Transl Psychiatry. 2020;10:160.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Li Y, Dong Y, Yang L, Tucker L, Yang B, Zong X, et al. Transcranial photobiomodulation prevents PTSD-like comorbidities in rats experiencing underwater trauma. Transl Psychiatry. 2021;11:270.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Askalsky P, Iosifescu DV. Transcranial photobiomodulation for the management of depression: current perspectives. Neuropsychiatr Dis Treat. 2019;15:3255–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Caldieraro MA, Sani G, Bui E, Cassano P. Long-term near-infrared photobiomodulation for anxious depression complicated by Takotsubo cardiomyopathy. J Clin Psychopharmacol. 2018;38:268–70.

    Article  PubMed  Google Scholar 

  118. Salehpour F, Hamblin MR, DiDuro JO. Rapid reversal of cognitive decline, olfactory dysfunction, and quality of life using multi-modality photobiomodulation therapy: case report. Photobiomodul Photomed Laser Surg. 2019;37:159–67.

    Article  PubMed  Google Scholar 

  119. Yang L, Dong Y, Wu C, Li Y, Guo Y, Yang B, et al. Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia-ischemia. J Biophotonics. 2019;12:e201800359.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Hamblin MR. Mechanisms and mitochondrial redox signaling in photobiomodulation. Photochem Photobiol. 2018;94:199–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Yang L, Tucker D, Dong Y, Wu C, Lu Y, Li Y, et al. Photobiomodulation therapy promotes neurogenesis by improving post-stroke local microenvironment and stimulating neuroprogenitor cells. Exp Neurol. 2018;299:86–96.

    Article  CAS  PubMed  Google Scholar 

  122. Poyton RO, Ball KA. Therapeutic photobiomodulation: nitric oxide and a novel function of mitochondrial cytochrome c oxidase. Discov Med. 2011;11:154–9.

    PubMed  Google Scholar 

  123. de Freitas LF, Hamblin MR. Proposed mechanisms of photobiomodulation or low-level light therapy. IEEE J Sel Top Quantum Electron. 2016;22:7000417.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Lane N. Cell biology: power games. Nature. 2006;443:901–3.

    Article  CAS  PubMed  Google Scholar 

  125. Bathini M, Raghushaker CR, Mahato KK. The molecular mechanisms of action of photobiomodulation against neurodegenerative diseases: a systematic review. Cell Mol Neurobiol. 2020;42(4):955–71.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Tatmatsu-Rocha JC, Tim CR, Avo L, Bernardes-Filho R, Brassolatti P, Kido HW, et al. Mitochondrial dynamics (fission and fusion) and collagen production in a rat model of diabetic wound healing treated by photobiomodulation: comparison of 904nm laser and 850nm light-emitting diode (LED). J Photochem Photobiol B. 2018;187:41–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Yu W, Wang X, Zhao J, Liu R, Liu J, Wang Z, et al. Stat2-Drp1 mediated mitochondrial mass increase is necessary for pro-inflammatory differentiation of macrophages. Redox Biol. 2020;37:101761.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Bonda DJ, Wang X, Perry G, Smith MA, Zhu X. Mitochondrial dynamics in Alzheimer’s disease: opportunities for future treatment strategies. Drugs Aging. 2010;27:181–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Foo ASC, Soong TW, Yeo TT, Lim KL. Mitochondrial dysfunction and Parkinson’s disease-near-infrared photobiomodulation as a potential therapeutic strategy. Front Aging Neurosci. 2020;12:89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wang R, Dong Y, Lu Y, Zhang W, Brann DW, Zhang Q. Photobiomodulation for global cerebral ischemia: targeting mitochondrial dynamics and functions. Mol Neurobiol. 2019;56:1852–69.

    Article  CAS  PubMed  Google Scholar 

  131. Song JW, Li K, Liang ZW, Dai C, Shen XF, Gong YZ, et al. Low-level laser facilitates alternatively activated macrophage/microglia polarization and promotes functional recovery after crush spinal cord injury in rats. Sci Rep. 2017;7:620.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. di Penta A, Moreno B, Reix S, Fernandez-Diez B, Villanueva M, Errea O, et al. Oxidative stress and proinflammatory cytokines contribute to demyelination and axonal damage in a cerebellar culture model of neuroinflammation. PLoS One. 2013;8:e54722.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Engert A, Schnell R, Lathan B, Diehl V. Limits and value of chemotherapy in elderly patients. Med Klin (Munich). 1994;89:603–10.

    CAS  PubMed  Google Scholar 

  134. Sunemi SM, Teixeira ILA, Mansano B, de Oliveira HA, Antonio EL, de Souza OC, et al. Post-resistance exercise photobiomodulation therapy has a more effective antioxidant effect than pre-application on muscle oxidative stress. Photochem Photobiol Sci. 2021;20:585–95.

    Article  CAS  PubMed  Google Scholar 

  135. de Oliveira HA, Antonio EL, Arsa G, Santana ET, Silva FA, Junior DA, et al. Photobiomodulation leads to reduced oxidative stress in rats submitted to high-intensity resistive exercise. Oxid Med Cell Longev. 2018;2018:5763256.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Dos Santos SA, Serra AJ, Stancker TG, Simoes MCB, Dos Santos Vieira MA, Leal-Junior EC, et al. Effects of photobiomodulation therapy on oxidative stress in muscle injury animal models: a systematic review. Oxid Med Cell Longev. 2017;2017:5273403.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Pratico D. Oxidative stress hypothesis in Alzheimer’s disease: a reappraisal. Trends Pharmacol Sci. 2008;29:609–15.

    Article  CAS  PubMed  Google Scholar 

  138. Bradley MA, Xiong-Fister S, Markesbery WR, Lovell MA. Elevated 4-hydroxyhexenal in Alzheimer’s disease (AD) progression. Neurobiol Aging. 2012;33:1034–44.

    Article  CAS  PubMed  Google Scholar 

  139. Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol. 1994;36:747–51.

    Article  CAS  PubMed  Google Scholar 

  140. Jiang H, Niu F, Zheng Y, Xu Y. CART mitigates oxidative stress and DNA damage in memory deficits of APP/PS1 mice via upregulating betaamyloid metabolismassociated enzymes. Mol Med Rep. 2021;23:280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Butterfield DA, Hensley K, Cole P, Subramaniam R, Aksenov M, Aksenova M, et al. Oxidatively induced structural alteration of glutamine synthetase assessed by analysis of spin label incorporation kinetics: relevance to Alzheimer’s disease. J Neurochem. 1997;68:2451–7.

    Article  CAS  PubMed  Google Scholar 

  142. Heo JC, Park JA, Kim DK, Lee JH. Photobiomodulation (660 nm) therapy reduces oxidative stress and induces BDNF expression in the hippocampus. Sci Rep. 2019;9:10114.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Purushothuman S, Johnstone DM, Nandasena C, Mitrofanis J, Stone J. Photobiomodulation with near infrared light mitigates Alzheimer’s disease-related pathology in cerebral cortex - evidence from two transgenic mouse models. Alzheimers Res Ther. 2014;6:2.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Zhang J, Wang X, Vikash V, Ye Q, Wu D, Liu Y, et al. ROS and ROS-mediated cellular signaling. Oxid Med Cell Longev. 2016;2016:4350965.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Chen AC, Arany PR, Huang YY, Tomkinson EM, Sharma SK, Kharkwal GB, et al. Low-level laser therapy activates NF-kB via generation of reactive oxygen species in mouse embryonic fibroblasts. PLoS One. 2011;6:e22453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Barolet D, Christiaens F, Hamblin MR. Infrared and skin: friend or foe. J Photochem Photobiol B. 2016;155:78–85.

    Article  CAS  PubMed  Google Scholar 

  147. Huang YY, Nagata K, Tedford CE, McCarthy T, Hamblin MR. Low-level laser therapy (LLLT) reduces oxidative stress in primary cortical neurons in vitro. J Biophotonics. 2013;6:829–38.

    CAS  PubMed  Google Scholar 

  148. Sommer AP. Mitochondrial cytochrome c oxidase is not the primary acceptor for near infrared light-it is mitochondrial bound water: the principles of low-level light therapy. Ann Transl Med. 2019;7:S13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Lima PLV, Pereira CV, Nissanka N, Arguello T, Gavini G, Maranduba C, et al. Photobiomodulation enhancement of cell proliferation at 660nm does not require cytochrome c oxidase. J Photochem Photobiol B. 2019;194:71–5.

    Article  CAS  PubMed  Google Scholar 

  150. Johnstone DM, Hamilton C, Gordon LC, Moro C, Torres N, Nicklason F, et al. Exploring the use of intracranial and extracranial (remote) photobiomodulation devices in Parkinson’s disease: a comparison of direct and indirect systemic stimulations. J Alzheimers Dis. 2021;83:1399–413.

    Article  PubMed  Google Scholar 

  151. Gordon LC, Johnstone DM. Remote photobiomodulation: an emerging strategy for neuroprotection. Neural Regen Res. 2019;14:2086–7.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Holczer A, Nemeth VL, Vekony T, Vecsei L, Klivenyi P, Must A. Non-invasive brain stimulation in Alzheimer’s disease and mild cognitive impairment-a state-of-the-art review on methodological characteristics and stimulation parameters. Front Hum Neurosci. 2020;14:179.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Weiler M, Stieger KC, Long JM, Rapp PR. Transcranial magnetic stimulation in Alzheimer’s disease: are we ready? eNeuro. 2020;7.ENEURO.0235-19.2019.

  154. Peng Z, Zhou C, Xue S, Bai J, Yu S, Li X, et al. Mechanism of repetitive transcranial magnetic stimulation for depression. Shanghai Arch Psychiatry. 2018;30:84–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Senda D, Strong H, Hines D, Hines R, Baker RJ. A compact 1200 V, 700 A, IGBT-based pulse generator for repetitive transcranial magnetic stimulation in vivo laboratory experiments on small animals. Rev Sci Instrum. 2021;92:084710.

    Article  CAS  PubMed  Google Scholar 

  156. Iglesias AH. Transcranial magnetic stimulation as treatment in multiple neurologic conditions. Curr Neurol Neurosci Rep. 2020;20:1.

    Article  PubMed  Google Scholar 

  157. Kombos T, Suss O. Neurophysiological basis of direct cortical stimulation and applied neuroanatomy of the motor cortex: a review. Neurosurg Focus. 2009;27:E3.

    Article  PubMed  Google Scholar 

  158. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic stimulation of human motor cortex. Lancet. 1985;1:1106–7.

    Article  CAS  PubMed  Google Scholar 

  159. Janicak PG, O'Reardon JP, Sampson SM, Husain MM, Lisanby SH, Rado JT, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222–32.

    Article  PubMed  Google Scholar 

  160. McClintock SM, Reti IM, Carpenter LL, McDonald WM, Dubin M, Taylor SF, et al. Consensus recommendations for the clinical application of repetitive transcranial magnetic stimulation (rTMS) in the treatment of depression. J Clin Psychiatry. 2018;79.16cs10905.

  161. George MS, Lisanby SH, Avery D, McDonald WM, Durkalski V, Pavlicova M, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham-controlled randomized trial. Arch Gen Psychiatry. 2010;67:507–16.

    Article  PubMed  Google Scholar 

  162. Levinson AJ, Fitzgerald PB, Favalli G, Blumberger DM, Daigle M, Daskalakis ZJ. Evidence of cortical inhibitory deficits in major depressive disorder. Biol Psychiatry. 2010;67:458–64.

    Article  CAS  PubMed  Google Scholar 

  163. Pohar R, Farrah K. In Repetitive transcranial magnetic stimulation for patients with depression: a review of clinical effectiveness, cost-effectiveness and guidelines - an update. Ottawa: CADTH Rapid Response Reports; 2019.

    Google Scholar 

  164. Health Quality O. Repetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and meta-analysis of randomized controlled trials. Ont Health Technol Assess Ser. 2016;16:1–66.

    Google Scholar 

  165. Ni Z, Chen R. Repetitive transcranial magnetic stimulation: faster or longer is not necessarily more. J Physiol. 2008;586:3733–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Klomjai W, Katz R, Lackmy-Vallee A. Basic principles of transcranial magnetic stimulation (TMS) and repetitive TMS (rTMS). Ann Phys Rehabil Med. 2015;58:208–13.

    Article  PubMed  Google Scholar 

  167. Drumond Marra HL, Myczkowski ML, Maia Memoria C, Arnaut D, Leite Ribeiro P, Sardinha Mansur CG, et al. Transcranial magnetic stimulation to address mild cognitive impairment in the elderly: a randomized controlled study. Behav Neurol. 2015;2015:287843.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Cotelli M, Manenti R, Cappa SF, Zanetti O, Miniussi C. Transcranial magnetic stimulation improves naming in Alzheimer disease patients at different stages of cognitive decline. Eur J Neurol. 2008;15:1286–92.

    Article  CAS  PubMed  Google Scholar 

  169. Di Lorenzo F, Bonni S, Picazio S, Motta C, Caltagirone C, Martorana A, et al. Effects of cerebellar theta burst stimulation on contralateral motor cortex excitability in patients with Alzheimer’s disease. Brain Topogr. 2020;33:613–7.

    Article  PubMed  Google Scholar 

  170. Hao Z, Wang D, Zeng Y, Liu M. Repetitive transcranial magnetic stimulation for improving function after stroke. Cochrane Database Syst Rev. 2013(5):CD008862.

  171. Lin Y, Jin J, Lv R, Luo Y, Dai W, Li W, et al. Repetitive transcranial magnetic stimulation increases the brain's drainage efficiency in a mouse model of Alzheimer’s disease. Acta Neuropathol Commun. 2021;9:102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Rajji TK. Transcranial magnetic and electrical stimulation in Alzheimer’s disease and mild cognitive impairment: a review of randomized controlled trials. Clin Pharmacol Ther. 2019;106:776–80.

    Article  PubMed  Google Scholar 

  173. Tatti E, Rossi S, Innocenti I, Rossi A, Santarnecchi E. Non-invasive brain stimulation of the aging brain: state of the art and future perspectives. Ageing Res Rev. 2016;29:66–89.

    Article  PubMed  Google Scholar 

  174. Chen X, Dong GY, Wang LX. High-frequency transcranial magnetic stimulation protects APP/PS1 mice against Alzheimer’s disease progress by reducing APOE and enhancing autophagy. Brain Behav. 2020;10:e01740.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Cotelli M, Calabria M, Manenti R, Rosini S, Zanetti O, Cappa SF, et al. Improved language performance in Alzheimer disease following brain stimulation. J Neurol Neurosurg Psychiatry. 2011;82:794–7.

    Article  PubMed  Google Scholar 

  176. Pang Y, Shi M. Repetitive transcranial magnetic stimulation improves mild cognitive impairment associated with Alzheimer’s disease in mice by modulating the miR-567/NEUROD2/PSD95 axis. Neuropsychiatr Dis Treat. 2021;17:2151–61.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Choung JS, Kim JM, Ko MH, Cho DS, Kim M. Therapeutic efficacy of repetitive transcranial magnetic stimulation in an animal model of Alzheimer’s disease. Sci Rep. 2021;11:437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Ma J, Zhang Z, Kang L, Geng D, Wang Y, Wang M, et al. Repetitive transcranial magnetic stimulation (rTMS) influences spatial cognition and modulates hippocampal structural synaptic plasticity in aging mice. Exp Gerontol. 2014;58:256–68.

    Article  PubMed  Google Scholar 

  179. Zhao J, Li Z, Cong Y, Zhang J, Tan M, Zhang H, et al. Repetitive transcranial magnetic stimulation improves cognitive function of Alzheimer’s disease patients. Oncotarget. 2017;8:33864–71.

    Article  PubMed  Google Scholar 

  180. Reithler J, Peters JC, Sack AT. Multimodal transcranial magnetic stimulation: using concurrent neuroimaging to reveal the neural network dynamics of noninvasive brain stimulation. Prog Neurobiol. 2011;94:149–65.

    Article  CAS  PubMed  Google Scholar 

  181. Tan T, Xie J, Tong Z, Liu T, Chen X, Tian X. Repetitive transcranial magnetic stimulation increases excitability of hippocampal CA1 pyramidal neurons. Brain Res. 2013;1520:23–35.

    Article  CAS  PubMed  Google Scholar 

  182. Wang F, Geng X, Tao HY, Cheng Y. The restoration after repetitive transcranial magnetic stimulation treatment on cognitive ability of vascular dementia rats and its impacts on synaptic plasticity in hippocampal CA1 area. J Mol Neurosci. 2010;41:145–55.

    Article  CAS  PubMed  Google Scholar 

  183. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med. 2011;1:a006189.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Moreno-Jimenez EP, Flor-Garcia M, Terreros-Roncal J, Rabano A, Cafini F, Pallas-Bazarra N, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25:554–60.

    Article  CAS  PubMed  Google Scholar 

  185. Caglayan AB, Beker MC, Caglayan B, Yalcin E, Caglayan A, Yulug B, et al. Acute and post-acute neuromodulation induces stroke recovery by promoting survival signaling, neurogenesis, and pyramidal tract plasticity. Front Cell Neurosci. 2019;13:144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Zhen J, Qian Y, Fu J, Su R, An H, Wang W, et al. Deep brain magnetic stimulation promotes neurogenesis and restores cholinergic activity in a transgenic mouse model of Alzheimer’s disease. Front Neural Circuits. 2017;11:48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Czeh B, Welt T, Fischer AK, Erhardt A, Schmitt W, Muller MB, et al. Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis. Biol Psychiatry. 2002;52:1057–65.

    Article  CAS  PubMed  Google Scholar 

  188. Heath A, Lindberg DR, Makowiecki K, Gray A, Asp AJ, Rodger J, et al. Medium- and high-intensity rTMS reduces psychomotor agitation with distinct neurobiologic mechanisms. Transl Psychiatry. 2018;8:126.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Zuo C, Cao H, Ding F, Zhao J, Huang Y, Li G, et al. Neuroprotective efficacy of different levels of high-frequency repetitive transcranial magnetic stimulation in mice with CUMS-induced depression: Involvement of the p11/BDNF/Homer1a signaling pathway. J Psychiatr Res. 2020;125:152–63.

    Article  PubMed  Google Scholar 

  190. He Y, Sun N, Wang Z, Zou W. Effect of repetitive transcranial magnetic stimulation (rTMS) for insomnia: a protocol for a systematic review. BMJ Open. 2019;9:e029206.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Luo J, Zheng H, Zhang L, Zhang Q, Li L, Pei Z, et al. High-frequency repetitive transcranial magnetic stimulation (rTMS) improves functional recovery by enhancing neurogenesis and activating BDNF/TrkB signaling in ischemic rats. Int J Mol Sci. 2017;18:455.

    Article  PubMed Central  CAS  Google Scholar 

  192. Pentkowski NS, Berkowitz LE, Thompson SM, Drake EN, Olguin CR, Clark BJ. Anxiety-like behavior as an early endophenotype in the TgF344-AD rat model of Alzheimer’s disease. Neurobiol Aging. 2018;61:169–76.

    Article  CAS  PubMed  Google Scholar 

  193. Ringman JM, Liang LJ, Zhou Y, Vangala S, Teng E, Kremen S, et al. Early behavioural changes in familial Alzheimer’s disease in the dominantly inherited Alzheimer network. Brain. 2015;138:1036–45.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Teixeira CM, Rosen ZB, Suri D, Sun Q, Hersh M, Sargin D, et al. Hippocampal 5-HT input regulates memory formation and Schaffer collateral excitation. Neuron. 2018;98:992–1004 e1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Ogren SO, Eriksson TM, Elvander-Tottie E, D'Addario C, Ekstrom JC, Svenningsson P, et al. The role of 5-HT(1A) receptors in learning and memory. Behav Brain Res. 2008;195:54–77.

    Article  PubMed  CAS  Google Scholar 

  196. Smith GS, Barrett FS, Joo JH, Nassery N, Savonenko A, Sodums DJ, et al. Molecular imaging of serotonin degeneration in mild cognitive impairment. Neurobiol Dis. 2017;105:33–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Christensen R, Marcussen AB, Wortwein G, Knudsen GM, Aznar S. Abeta(1-42) injection causes memory impairment, lowered cortical and serum BDNF levels, and decreased hippocampal 5-HT(2A) levels. Exp Neurol. 2008;210:164–71.

    Article  CAS  PubMed  Google Scholar 

  198. Thomas AJ, Hendriksen M, Piggott M, Ferrier IN, Perry E, Ince P, et al. A study of the serotonin transporter in the prefrontal cortex in late-life depression and Alzheimer’s disease with and without depression. Neuropathol Appl Neurobiol. 2006;32:296–303.

    Article  CAS  PubMed  Google Scholar 

  199. Bouaziz N, Laidi C, Thomas F, Schenin-King Andrianisaina P, Moulier V, Januel D. Intermittent theta-burst stimulation over the dorsolateral prefrontal cortex (DLPFC) in healthy subjects produces no cumulative effect on cortical excitability. Front Psychiatry. 2021;12:626479.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Lefaucheur JP, Aleman A, Baeken C, Benninger DH, Brunelin J, Di Lazzaro V, et al. Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS): an update (2014-2018). Clin Neurophysiol. 2020;131:474–528.

    Article  PubMed  Google Scholar 

  201. Askin A, Tosun A, Demirdal US. Effects of low-frequency repetitive transcranial magnetic stimulation on upper extremity motor recovery and functional outcomes in chronic stroke patients: a randomized controlled trial. Somatosens Mot Res. 2017;34:102–7.

    Article  PubMed  Google Scholar 

  202. Baeken C, De Raedt R, Bossuyt A, Van Hove C, Mertens J, Dobbeleir A, et al. The impact of HF-rTMS treatment on serotonin(2A) receptors in unipolar melancholic depression. Brain Stimul. 2011;4:104–11.

    Article  PubMed  Google Scholar 

  203. Chervyakov AV, Chernyavsky AY, Sinitsyn DO, Piradov MA. Possible mechanisms underlying the therapeutic effects of transcranial magnetic stimulation. Front Hum Neurosci. 2015;9:303.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Miniussi C, Bonato C, Bignotti S, Gazzoli A, Gennarelli M, Pasqualetti P, et al. Repetitive transcranial magnetic stimulation (rTMS) at high and low frequency: an efficacious therapy for major drug-resistant depression? Clin Neurophysiol. 2005;116:1062–71.

    Article  CAS  PubMed  Google Scholar 

  205. Cannizzaro C, Talani G, Brancato A, Mulas G, Spiga S, De Luca MA, et al. Dopamine restores limbic memory loss, dendritic spine structure, and NMDAR-dependent LTD in the nucleus accumbens of alcohol-withdrawn rats. J Neurosci. 2019;39:929–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Clos M, Bunzeck N, Sommer T. Dopamine is a double-edged sword: dopaminergic modulation enhances memory retrieval performance but impairs metacognition. Neuropsychopharmacology. 2019;44:555–63.

    Article  CAS  PubMed  Google Scholar 

  207. Gonzalez-Burgos I, Feria-Velasco A. Serotonin/dopamine interaction in memory formation. Prog Brain Res. 2008;172:603–23.

    Article  CAS  PubMed  Google Scholar 

  208. Pogarell O, Koch W, Popperl G, Tatsch K, Jakob F, Zwanzger P, et al. Striatal dopamine release after prefrontal repetitive transcranial magnetic stimulation in major depression: preliminary results of a dynamic [123I] IBZM SPECT study. J Psychiatr Res. 2006;40:307–14.

    Article  PubMed  Google Scholar 

  209. Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, et al. Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci U S A. 2010;107:2580–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Solas M, Puerta E, Ramirez MJ. Treatment options in Alzheimer’s disease: the GABA story. Curr Pharm Des. 2015;21:4960–71.

    Article  CAS  PubMed  Google Scholar 

  211. Ren Z, Sahir N, Murakami S, Luellen BA, Earnheart JC, Lal R, et al. Defects in dendrite and spine maturation and synaptogenesis associated with an anxious-depressive-like phenotype of GABAA receptor-deficient mice. Neuropharmacology. 2015;88:171–9.

    Article  CAS  PubMed  Google Scholar 

  212. Dubin MJ, Mao X, Banerjee S, Goodman Z, Lapidus KA, Kang G, et al. Elevated prefrontal cortex GABA in patients with major depressive disorder after TMS treatment measured with proton magnetic resonance spectroscopy. J Psychiatry Neurosci. 2016;41:E37–45.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Wang H, Geng Y, Han B, Qiang J, Li X, Sun M, et al. Repetitive transcranial magnetic stimulation applications normalized prefrontal dysfunctions and cognitive-related metabolic profiling in aged mice. PLoS One. 2013;8:e81482.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Aftanas LI, Gevorgyan MM, Zhanaeva SY, Dzemidovich SS, Kulikova KI, Al'perina EL, et al. Therapeutic effects of repetitive transcranial magnetic stimulation (rTMS) on neuroinflammation and neuroplasticity in patients with Parkinson’s disease: a placebo-controlled study. Bull Exp Biol Med. 2018;165:195–9.

    Article  CAS  PubMed  Google Scholar 

  215. Sasso V, Bisicchia E, Latini L, Ghiglieri V, Cacace F, Carola V, et al. Repetitive transcranial magnetic stimulation reduces remote apoptotic cell death and inflammation after focal brain injury. J Neuroinflammation. 2016;13:150.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Hong Y, Liu Q, Peng M, Bai M, Li J, Sun R, et al. High-frequency repetitive transcranial magnetic stimulation improves functional recovery by inhibiting neurotoxic polarization of astrocytes in ischemic rats. J Neuroinflammation. 2020;17:150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Luan D, Zhao MG, Shi YC, Li L, Cao YJ, Feng HX, et al. Mechanisms of repetitive transcranial magnetic stimulation for anti-depression: evidence from preclinical studies. World J Psychiatry. 2020;10:223–33.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Wang HN, Bai YH, Chen YC, Zhang RG, Wang HH, Zhang YH, et al. Repetitive transcranial magnetic stimulation ameliorates anxiety-like behavior and impaired sensorimotor gating in a rat model of post-traumatic stress disorder. PLoS One. 2015;10:e0117189.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Li K, Wang X, Jiang Y, Zhang X, Liu Z, Yin T, et al. Early intervention attenuates synaptic plasticity impairment and neuroinflammation in 5xFAD mice. J Psychiatr Res. 2021;136:204–16.

    Article  PubMed  Google Scholar 

  220. Clarke D, Beros J, Bates KA, Harvey AR, Tang AD, Rodger J. Low intensity repetitive magnetic stimulation reduces expression of genes related to inflammation and calcium signalling in cultured mouse cortical astrocytes. Brain Stimul. 2021;14:183–91.

    Article  PubMed  Google Scholar 

  221. Shigetomi E, Saito K, Sano F, Koizumi S. Aberrant calcium signals in reactive astrocytes: a key process in neurological disorders. Int J Mol Sci. 2019;20:996.

    Article  CAS  PubMed Central  Google Scholar 

  222. Chen SY, Gao Y, Sun JY, Meng XL, Yang D, Fan LH, et al. Traditional Chinese medicine: role in reducing beta-amyloid, apoptosis, autophagy, neuroinflammation, oxidative stress, and mitochondrial dysfunction of Alzheimer’s disease. Front Pharmacol. 2020;11:497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Velioglu HA, Hanoglu L, Bayraktaroglu Z, Toprak G, Guler EM, Bektay MY, et al. Left lateral parietal rTMS improves cognition and modulates resting brain connectivity in patients with Alzheimer’s disease: possible role of BDNF and oxidative stress. Neurobiol Learn Mem. 2021;180:107410.

    Article  CAS  PubMed  Google Scholar 

  224. Zong X, Li Y, Liu C, Qi W, Han D, Tucker L, et al. Theta-burst transcranial magnetic stimulation promotes stroke recovery by vascular protection and neovascularization. Theranostics. 2020;10:12090–110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Aguera E, Caballero-Villarraso J, Feijoo M, Escribano BM, Bahamonde MC, Conde C, et al. Impact of repetitive transcranial magnetic stimulation on neurocognition and oxidative stress in relapsing-remitting multiple sclerosis: A Case Report. Front Neurol. 2020;11:817.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Milev RV, Giacobbe P, Kennedy SH, Blumberger DM, Daskalakis ZJ, Downar J, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: section 4. Neurostimulation treatments. Can J Psychiatry. 2016;61:561–75.

    Article  PubMed  Google Scholar 

  227. Perera T, George MS, Grammer G, Janicak PG, Pascual-Leone A, Wirecki TS. The clinical TMS society consensus review and treatment recommendations for TMS therapy for major depressive disorder. Brain Stimul. 2016;9:336–46.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Durmaz O, Ispir E, Baykan H, Alisik M, Erel O. The impact of repetitive transcranial magnetic stimulation on oxidative stress in subjects with medication-resistant depression. J ECT. 2018;34:127–31.

    Article  PubMed  Google Scholar 

  229. Tunez I, Montilla P, del Carmen MM, Medina FJ, Drucker-Colin R. Effect of transcranial magnetic stimulation on oxidative stress induced by 3-nitropropionic acid in cortical synaptosomes. Neurosci Res. 2006;56:91–5.

    Article  CAS  PubMed  Google Scholar 

  230. Zhao L, Ren H, Gu S, Li X, Jiang C, Li J, et al. rTMS ameliorated depressive-like behaviors by restoring HPA axis balance and prohibiting hippocampal neuron apoptosis in a rat model of depression. Psychiatry Res. 2018;269:126–33.

    Article  CAS  PubMed  Google Scholar 

  231. Chen X, Chen S, Liang W, Ba F. Administration of repetitive transcranial magnetic stimulation attenuates abeta 1-42-induced Alzheimer’s disease in mice by activating beta-catenin signaling. Biomed Res Int. 2019;2019:1431760.

    PubMed  PubMed Central  Google Scholar 

  232. Rutherford G, Lithgow B, Moussavi Z. Short and long-term effects of rTMS treatment on Alzheimer’s disease at different stages: a pilot study. J Exp Neurosci. 2015;9:43–51.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Kumar S, Zomorrodi R, Ghazala Z, Goodman MS, Blumberger DM, Cheam A, et al. Extent of dorsolateral prefrontal cortex plasticity and its association with working memory in patients with Alzheimer disease. JAMA Psychiatry. 2017;74:1266–74.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Stultz DJ, Osburn S, Burns T, Pawlowska-Wajswol S, Walton R. Transcranial magnetic stimulation (TMS) safety with respect to seizures: a literature review. Neuropsychiatr Dis Treat. 2020;16:2989–3000.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Thair H, Holloway AL, Newport R, Smith AD. Transcranial direct current stimulation (tDCS): a beginner’s guide for design and implementation. Front Neurosci. 2017;11:641.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Bennabi D, Haffen E. Transcranial direct current stimulation (tDCS): a promising treatment for major depressive disorder? Brain Sci. 2018;8:81.

    Article  PubMed Central  CAS  Google Scholar 

  237. Stein DJ, Fernandes Medeiros L, Caumo W, Torres IL. Transcranial direct current stimulation in patients with anxiety: current perspectives. Neuropsychiatr Dis Treat. 2020;16:161–9.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Lee HK, Ahn SJ, Shin YM, Kang N, Cauraugh JH. Does transcranial direct current stimulation improve functional locomotion in people with Parkinson’s disease? A systematic review and meta-analysis. J Neuroeng Rehabil. 2019;16:84.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Zaninotto AL, El-Hagrassy MM, Green JR, Babo M, Paglioni VM, Benute GG, et al. Transcranial direct current stimulation (tDCS) effects on traumatic brain injury (TBI) recovery: a systematic review. Dement Neuropsychol. 2019;13:172–9.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Andrade SM, de Mendonca C, Pereira TCL, Fernandez-Calvo B, Araujo RCN, Alves NT. Adjuvant transcranial direct current stimulation for treating Alzheimer’s disease: a case study. Dement Neuropsychol. 2016;10:156–9.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Park J, Oh Y, Chung K, Kim KJ, Kim CO, Park JY. Effect of home-based transcranial direct current stimulation (tDCS) on cognitive function in patients with mild cognitive impairment: a study protocol for a randomized, double-blind, cross-over study. Trials. 2019;20:278.

    Article  PubMed  PubMed Central  Google Scholar 

  242. Khedr EM, Salama RH, Abdel Hameed M, Abo Elfetoh N, Seif P. Therapeutic role of transcranial direct current stimulation in Alzheimer disease patients: double-blind, placebo-controlled clinical trial. Neurorehabil Neural Repair. 2019;33:384–94.

    Article  PubMed  Google Scholar 

  243. Yang WJ, Wen HZ, Zhou LX, Luo YP, Hou WS, Wang X, et al. After-effects of repetitive anodal transcranial direct current stimulation on learning and memory in a rat model of Alzheimer’s disease. Neurobiol Learn Mem. 2019;161:37–45.

    Article  PubMed  Google Scholar 

  244. Nitsche MA, Paulus W. Excitability changes induced in the human motor cortex by weak transcranial direct current stimulation. J Physiol. 2000;527(Pt 3):633–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Zaehle T, Sandmann P, Thorne JD, Jancke L, Herrmann CS. Transcranial direct current stimulation of the prefrontal cortex modulates working memory performance: combined behavioural and electrophysiological evidence. BMC Neurosci. 2011;12:2.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Fregni F, Boggio PS, Nitsche M, Bermpohl F, Antal A, Feredoes E, et al. Anodal transcranial direct current stimulation of prefrontal cortex enhances working memory. Exp Brain Res. 2005;166:23–30.

    Article  PubMed  Google Scholar 

  247. Ohn SH, Park CI, Yoo WK, Ko MH, Choi KP, Kim GM, et al. Time-dependent effect of transcranial direct current stimulation on the enhancement of working memory. Neuroreport. 2008;19:43–7.

    Article  PubMed  Google Scholar 

  248. Boggio PS, Ferrucci R, Mameli F, Martins D, Martins O, Vergari M, et al. Prolonged visual memory enhancement after direct current stimulation in Alzheimer’s disease. Brain Stimul. 2012;5:223–30.

    Article  PubMed  Google Scholar 

  249. Ferrucci R, Mameli F, Guidi I, Mrakic-Sposta S, Vergari M, Marceglia S, et al. Transcranial direct current stimulation improves recognition memory in Alzheimer disease. Neurology. 2008;71:493–8.

    Article  CAS  PubMed  Google Scholar 

  250. Gangemi A, Colombo B, Fabio RA. Effects of short- and long-term neurostimulation (tDCS) on Alzheimer’s disease patients: two randomized studies. Aging Clin Exp Res. 2021;33:383–90.

    Article  PubMed  Google Scholar 

  251. Luo Y, Yang W, Li N, Yang X, Zhu B, Wang C, et al. Anodal transcranial direct current stimulation can improve spatial learning and memory and attenuate Abeta42 burden at the early stage of Alzheimer’s disease in APP/PS1 transgenic mice. Front Aging Neurosci. 2020;12:134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Luo Y, Yang H, Yan X, Wu Y, Wei G, Wu X, et al. Transcranial direct current stimulation alleviates neurovascular unit dysfunction in mice with preclinical Alzheimer’s disease. Front Aging Neurosci. 2022;14:857415.

    Article  PubMed  PubMed Central  Google Scholar 

  253. Wolters FJ, Zonneveld HI, Hofman A, van der Lugt A, Koudstaal PJ, Vernooij MW, et al. Cerebral perfusion and the risk of dementia: a population-based study. Circulation. 2017;136:719–28.

    Article  PubMed  Google Scholar 

  254. Roher AE, Debbins JP, Malek-Ahmadi M, Chen K, Pipe JG, Maze S, et al. Cerebral blood flow in Alzheimer’s disease. Vasc Health Risk Manag. 2012;8:599–611.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Korte N, Nortley R, Attwell D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer’s disease. Acta Neuropathol. 2020;140:793–810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Benedictus MR, Leeuwis AE, Binnewijzend MA, Kuijer JP, Scheltens P, Barkhof F, et al. Lower cerebral blood flow is associated with faster cognitive decline in Alzheimer’s disease. Eur Radiol. 2017;27:1169–75.

    Article  PubMed  Google Scholar 

  257. Mughal A, Harraz OF, Gonzales AL, Hill-Eubanks D, Nelson MT. PIP2 improves cerebral blood flow in a mouse model of Alzheimer’s disease. Function (Oxf). 2021;2:zqab010.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Bracko O, Njiru BN, Swallow M, Ali M, Haft-Javaherian M, Schaffer CB. Increasing cerebral blood flow improves cognition into late stages in Alzheimer’s disease mice. J Cereb Blood Flow Metab. 2020;40:1441–52.

    Article  CAS  PubMed  Google Scholar 

  259. de Jong DLK, de Heus RAA, Rijpma A, Donders R, Olde Rikkert MGM, Gunther M, et al. Effects of nilvadipine on cerebral blood flow in patients with Alzheimer disease. Hypertension. 2019;74:413–20.

    Article  PubMed  CAS  Google Scholar 

  260. Zheng X, Alsop DC, Schlaug G. Effects of transcranial direct current stimulation (tDCS) on human regional cerebral blood flow. Neuroimage. 2011;58:26–33.

    Article  PubMed  Google Scholar 

  261. Hu S, Zheng T, Dong Y, Du J, Liu L. Effect of anodal direct-current stimulation on cortical hemodynamic responses with laser-speckle contrast imaging. Front Neurosci. 2018;12:503.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Merzagora AC, Foffani G, Panyavin I, Mordillo-Mateos L, Aguilar J, Onaral B, et al. Prefrontal hemodynamic changes produced by anodal direct current stimulation. Neuroimage. 2010;49:2304–10.

    Article  CAS  PubMed  Google Scholar 

  263. Citri A, Malenka RC. Synaptic plasticity: multiple forms, functions, and mechanisms. Neuropsychopharmacology. 2008;33:18–41.

    Article  PubMed  Google Scholar 

  264. Benarroch EE. Glutamatergic synaptic plasticity and dysfunction in Alzheimer disease: emerging mechanisms. Neurology. 2018;91:125–32.

    Article  PubMed  Google Scholar 

  265. Yuan TF, Li WG, Zhang C, Wei H, Sun S, Xu NJ, et al. Targeting neuroplasticity in patients with neurodegenerative diseases using brain stimulation techniques. Transl Neurodegener. 2020;9:44.

    Article  PubMed  PubMed Central  Google Scholar 

  266. Nitsche MA, Fricke K, Henschke U, Schlitterlau A, Liebetanz D, Lang N, et al. Pharmacological modulation of cortical excitability shifts induced by transcranial direct current stimulation in humans. J Physiol. 2003;553:293–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Stagg CJ, Nitsche MA. Physiological basis of transcranial direct current stimulation. Neuroscientist. 2011;17:37–53.

    Article  PubMed  Google Scholar 

  268. Lisman JE. Three Ca2+ levels affect plasticity differently: the LTP zone, the LTD zone and no man’s land. J Physiol. 2001;532:285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Oda Y. Choline acetyltransferase: the structure, distribution and pathologic changes in the central nervous system. Pathol Int. 1999;49:921–37.

    Article  CAS  PubMed  Google Scholar 

  270. Cerpa W, Dinamarca MC, Inestrosa NC. Structure-function implications in Alzheimer’s disease: effect of Abeta oligomers at central synapses. Curr Alzheimer Res. 2008;5:233–43.

    Article  CAS  PubMed  Google Scholar 

  271. Stagg CJ, Best JG, Stephenson MC, O'Shea J, Wylezinska M, Kincses ZT, et al. Polarity-sensitive modulation of cortical neurotransmitters by transcranial stimulation. J Neurosci. 2009;29:5202–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Thirugnanasambandam N, Grundey J, Adam K, Drees A, Skwirba AC, Lang N, et al. Nicotinergic impact on focal and non-focal neuroplasticity induced by non-invasive brain stimulation in non-smoking humans. Neuropsychopharmacology. 2011;36:879–86.

    Article  CAS  PubMed  Google Scholar 

  273. Monte-Silva K, Liebetanz D, Grundey J, Paulus W, Nitsche MA. Dosage-dependent non-linear effect of L-dopa on human motor cortex plasticity. J Physiol. 2010;588:3415–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Nitsche MA, Lampe C, Antal A, Liebetanz D, Lang N, Tergau F, et al. Dopaminergic modulation of long-lasting direct current-induced cortical excitability changes in the human motor cortex. Eur J Neurosci. 2006;23:1651–7.

    Article  PubMed  Google Scholar 

  275. Guo T, Fang J, Tong ZY, He S, Luo Y. Transcranial direct current stimulation ameliorates cognitive impairment via modulating oxidative stress, inflammation, and autophagy in a rat model of vascular dementia. Front Neurosci. 2020;14:28.

    Article  PubMed  PubMed Central  Google Scholar 

  276. Rueger MA, Keuters MH, Walberer M, Braun R, Klein R, Sparing R, et al. Multi-session transcranial direct current stimulation (tDCS) elicits inflammatory and regenerative processes in the rat brain. PLoS One. 2012;7:e43776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Peruzzotti-Jametti L, Cambiaghi M, Bacigaluppi M, Gallizioli M, Gaude E, Mari S, et al. Safety and efficacy of transcranial direct current stimulation in acute experimental ischemic stroke. Stroke. 2013;44:3166–74.

    Article  PubMed  Google Scholar 

  278. Vulic K, Bjekic J, Paunovic D, Jovanovic M, Milanovic S, Filipovic SR. Theta-modulated oscillatory transcranial direct current stimulation over posterior parietal cortex improves associative memory. Sci Rep. 2021;11:3013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Accardi A, Wornell G. Quasi light fields: extending the light field to coherent radiation. J Opt Soc Am A Opt Image Sci Vis. 2009;26:2055–66.

    Article  PubMed  Google Scholar 

  280. Lee SB, Youn J, Jang W, Yang HO. Neuroprotective effect of anodal transcranial direct current stimulation on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in mice through modulating mitochondrial dynamics. Neurochem Int. 2019;129:104491.

    Article  CAS  PubMed  Google Scholar 

  281. Cui MY, Lin Y, Sheng JY, Zhang X, Cui RJ. Exercise intervention associated with cognitive improvement in Alzheimer’s disease. Neural Plast. 2018;2018:9234105.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  282. Meng Q, Lin MS, Tzeng IS. Relationship between exercise and Alzheimer’s disease: a narrative literature review. Front Neurosci. 2020;14:131.

    Article  PubMed  PubMed Central  Google Scholar 

  283. Nascimento SL, Surita FG, Cecatti JG. Physical exercise during pregnancy: a systematic review. Curr Opin Obstet Gynecol. 2012;24:387–94.

    Article  PubMed  Google Scholar 

  284. Di Donato DM, West DW, Churchward-Venne TA, Breen L, Baker SK, Phillips SM. Influence of aerobic exercise intensity on myofibrillar and mitochondrial protein synthesis in young men during early and late postexercise recovery. Am J Physiol Endocrinol Metab. 2014;306:E1025–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  285. Cordes T, Bischoff LL, Schoene D, Schott N, Voelcker-Rehage C, Meixner C, et al. A multicomponent exercise intervention to improve physical functioning, cognition and psychosocial well-being in elderly nursing home residents: a study protocol of a randomized controlled trial in the PROCARE (prevention and occupational health in long-term care) project. BMC Geriatr. 2019;19:369.

    Article  PubMed  PubMed Central  Google Scholar 

  286. Lu Y, Dong Y, Tucker D, Wang R, Ahmed ME, Brann D, et al. Treadmill exercise exerts neuroprotection and regulates microglial polarization and oxidative stress in a streptozotocin-induced rat model of sporadic Alzheimer’s disease. J Alzheimers Dis. 2017;56:1469–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Belarbi K, Burnouf S, Fernandez-Gomez FJ, Laurent C, Lestavel S, Figeac M, et al. Beneficial effects of exercise in a transgenic mouse model of Alzheimer’s disease-like Tau pathology. Neurobiol Dis. 2011;43:486–94.

    Article  CAS  PubMed  Google Scholar 

  288. Zhang X, He Q, Huang T, Zhao N, Liang F, Xu B, et al. Treadmill exercise decreases abeta deposition and counteracts cognitive decline in APP/PS1 mice, possibly via hippocampal microglia modifications. Front Aging Neurosci. 2019;11:78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Donovan NJ, Locascio JJ, Marshall GA, Gatchel J, Hanseeuw BJ, Rentz DM, et al. Longitudinal association of amyloid beta and anxious-depressive symptoms in cognitively normal older adults. Am J Psychiatry. 2018;175:530–7.

    Article  PubMed  PubMed Central  Google Scholar 

  290. Meng FT, Zhao J, Fang H, Liu YJ. The influence of chronic stress on anxiety-like behavior and cognitive function in different human GFAP-ApoE transgenic adult male mice. Stress. 2015;18:419–26.

    Article  PubMed  CAS  Google Scholar 

  291. Hsiao JJ, Teng E. Depressive symptoms in clinical and incipient Alzheimer’s disease. Neurodegener Dis Manag. 2013;3:147–55.

    Article  PubMed  Google Scholar 

  292. Geda YE, Roberts RO, Knopman DS, Petersen RC, Christianson TJ, Pankratz VS, et al. Prevalence of neuropsychiatric symptoms in mild cognitive impairment and normal cognitive aging: population-based study. Arch Gen Psychiatry. 2008;65:1193–8.

    Article  PubMed  PubMed Central  Google Scholar 

  293. Zhang YL, Xing RZ, Luo XB, Xu H, Chang RC, Zou LY, et al. Anxiety-like behavior and dysregulation of miR-34a in triple transgenic mice of Alzheimer’s disease. Eur Rev Med Pharmacol Sci. 2016;20:2853–62.

    PubMed  Google Scholar 

  294. Williams CL, Tappen RM. Exercise training for depressed older adults with Alzheimer’s disease. Aging Ment Health. 2008;12:72–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. van der Kleij LA, Petersen ET, Siebner HR, Hendrikse J, Frederiksen KS, Sobol NA, et al. The effect of physical exercise on cerebral blood flow in Alzheimer’s disease. Neuroimage Clin. 2018;20:650–4.

    Article  PubMed  PubMed Central  Google Scholar 

  296. Austin BP, Nair VA, Meier TB, Xu G, Rowley HA, Carlsson CM, et al. Effects of hypoperfusion in Alzheimer’s disease. J Alzheimers Dis. 2011;26(Suppl 3):123–33.

    Article  PubMed  PubMed Central  Google Scholar 

  297. Binnewijzend MA, Kuijer JP, Benedictus MR, van der Flier WM, Wink AM, Wattjes MP, et al. Cerebral blood flow measured with 3D pseudocontinuous arterial spin-labeling MR imaging in Alzheimer disease and mild cognitive impairment: a marker for disease severity. Radiology. 2013;267:221–30.

    Article  PubMed  Google Scholar 

  298. Wolk DA, Detre JA. Arterial spin labeling MRI: an emerging biomarker for Alzheimer’s disease and other neurodegenerative conditions. Curr Opin Neurol. 2012;25:421–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Bangen KJ, Clark AL, Edmonds EC, Evangelista ND, Werhane ML, Thomas KR, et al. Cerebral blood flow and amyloid-beta interact to affect memory performance in cognitively normal older adults. Front Aging Neurosci. 2017;9:181.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  300. Mattsson N, Tosun D, Insel PS, Simonson A, Jack CR Jr, Beckett LA, et al. Association of brain amyloid-beta with cerebral perfusion and structure in Alzheimer's disease and mild cognitive impairment. Brain. 2014;137:1550–61.

    Article  PubMed  PubMed Central  Google Scholar 

  301. Bliss ES, Wong RH, Howe PR, Mills DE. Benefits of exercise training on cerebrovascular and cognitive function in ageing. J Cereb Blood Flow Metab. 2021;41:447–70.

    Article  PubMed  Google Scholar 

  302. De la Rosa A, Olaso-Gonzalez G, Arc-Chagnaud C, Millan F, Salvador-Pascual A, Garcia-Lucerga C, et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J Sport Health Sci. 2020;9:394–404.

    Article  PubMed  PubMed Central  Google Scholar 

  303. Globus M, Melamed E, Keren A, Tzivoni D, Granot C, Lavy S, et al. Effect of exercise on cerebral circulation. J Cereb Blood Flow Metab. 1983;3:287–90.

    Article  CAS  PubMed  Google Scholar 

  304. Scheinberg P, Blackburn LI, Rich M, Saslaw M. Effects of vigorous physical exercise on cerebral circulation and metabolism. Am J Med. 1954;16:549–54.

    Article  CAS  PubMed  Google Scholar 

  305. Smith KJ, Ainslie PN. Regulation of cerebral blood flow and metabolism during exercise. Exp Physiol. 2017;102:1356–71.

    Article  CAS  PubMed  Google Scholar 

  306. Imray CH, Walsh S, Clarke T, Tiivas C, Hoar H, Harvey TC, et al. Effects of breathing air containing 3% carbon dioxide, 35% oxygen or a mixture of 3% carbon dioxide/35% oxygen on cerebral and peripheral oxygenation at 150 m and 3459 m. Clin Sci (Lond). 2003;104:203–10.

    Article  CAS  PubMed  Google Scholar 

  307. Murphy MN, Mizuno M, Mitchell JH, Smith SA. Cardiovascular regulation by skeletal muscle reflexes in health and disease. Am J Physiol Heart Circ Physiol. 2011;301:H1191–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  308. Querido JS, Sheel AW. Regulation of cerebral blood flow during exercise. Sports Med. 2007;37:765–82.

    Article  PubMed  Google Scholar 

  309. Tomoto T, Liu J, Tseng BY, Pasha EP, Cardim D, Tarumi T, et al. One-year aerobic exercise reduced carotid arterial stiffness and increased cerebral blood flow in amnestic mild cognitive impairment. J Alzheimers Dis. 2021;80:841–53.

    Article  CAS  PubMed  Google Scholar 

  310. Jensen CS, Bahl JM, Ostergaard LB, Hogh P, Wermuth L, Heslegrave A, et al. Exercise as a potential modulator of inflammation in patients with Alzheimer’s disease measured in cerebrospinal fluid and plasma. Exp Gerontol. 2019;121:91–8.

    Article  CAS  PubMed  Google Scholar 

  311. Jiang T, Luo J, Pan X, Zheng H, Yang H, Zhang L, et al. Physical exercise modulates the astrocytes polarization, promotes myelin debris clearance and remyelination in chronic cerebral hypoperfusion rats. Life Sci. 2021;278:119526.

    Article  CAS  PubMed  Google Scholar 

  312. Valori CF, Possenti A, Brambilla L, Rossi D. Challenges and opportunities of targeting astrocytes to halt neurodegenerative disorders. Cells. 2021;10:2019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Belaya I, Ivanova M, Sorvari A, Ilicic M, Loppi S, Koivisto H, et al. Astrocyte remodeling in the beneficial effects of long-term voluntary exercise in Alzheimer’s disease. J Neuroinflammation. 2020;17:271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Garcia-Mesa Y, Colie S, Corpas R, Cristofol R, Comellas F, Nebreda AR, et al. Oxidative stress is a central target for physical exercise neuroprotection against pathological brain aging. J Gerontol A Biol Sci Med Sci. 2016;71:40–9.

    Article  PubMed  CAS  Google Scholar 

  315. Zhang H, Liu H, Davies KJ, Sioutas C, Finch CE, Morgan TE, et al. Nrf2-regulated phase II enzymes are induced by chronic ambient nanoparticle exposure in young mice with age-related impairments. Free Radic Biol Med. 2012;52:2038–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Mrakic-Sposta S, Di Santo SG, Franchini F, Arlati S, Zangiacomi A, Greci L, et al. Effects of combined physical and cognitive virtual reality-based training on cognitive impairment and oxidative stress in mci patients: a pilot study. Front Aging Neurosci. 2018;10:282.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  317. Simioni C, Zauli G, Martelli AM, Vitale M, Sacchetti G, Gonelli A, et al. Oxidative stress: role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget. 2018;9:17181–98.

    Article  PubMed  PubMed Central  Google Scholar 

  318. Natale G, Limanaqi F, Busceti CL, Mastroiacovo F, Nicoletti F, Puglisi-Allegra S, et al. Glymphatic system as a gateway to connect neurodegeneration from periphery to CNS. Front Neurosci. 2021;15:639140.

    Article  PubMed  PubMed Central  Google Scholar 

  319. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4:147ra111.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  320. Kaur J, Davoodi-Bojd E, Fahmy LM, Zhang L, Ding G, Hu J, et al. Magnetic resonance imaging and modeling of the glymphatic system. Diagnostics (Basel). 2020;10:344.

    Article  CAS  PubMed Central  Google Scholar 

  321. Boespflug EL, Iliff JJ. The emerging relationship between interstitial fluid-cerebrospinal fluid exchange, amyloid-beta, and sleep. Biol Psychiatry. 2018;83:328–36.

    Article  PubMed  Google Scholar 

  322. He XF, Liu DX, Zhang Q, Liang FY, Dai GY, Zeng JS, et al. Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci. 2017;10:144.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  323. Zeppenfeld DM, Simon M, Haswell JD, D'Abreo D, Murchison C, Quinn JF, et al. Association of perivascular localization of aquaporin-4 with cognition and Alzheimer disease in aging brains. JAMA Neurol. 2017;74:91–9.

    Article  PubMed  Google Scholar 

  324. Xu Z, Xiao N, Chen Y, Huang H, Marshall C, Gao J, et al. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Abeta accumulation and memory deficits. Mol Neurodegener. 2015;10:58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  325. von Holstein-Rathlou S, Petersen NC, Nedergaard M. Voluntary running enhances glymphatic influx in awake behaving, young mice. Neurosci Lett. 2018;662:253–8.

    Article  CAS  Google Scholar 

  326. Bernardo TC, Marques-Aleixo I, Beleza J, Oliveira PJ, Ascensao A, Magalhaes J. Physical exercise and brain mitochondrial fitness: the possible role against Alzheimer’s disease. Brain Pathol. 2016;26:648–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  327. Lanzillotta C, Di Domenico F, Perluigi M, Butterfield DA. Targeting mitochondria in Alzheimer disease: rationale and perspectives. CNS Drugs. 2019;33:957–69.

    Article  PubMed  PubMed Central  Google Scholar 

  328. Gusdon AM, Callio J, Distefano G, O'Doherty RM, Goodpaster BH, Coen PM, et al. Exercise increases mitochondrial complex I activity and DRP1 expression in the brains of aged mice. Exp Gerontol. 2017;90:1–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  329. Radak Z, Ihasz F, Koltai E, Goto S, Taylor AW, Boldogh I. The redox-associated adaptive response of brain to physical exercise. Free Radic Res. 2014;48:84–92.

    Article  CAS  PubMed  Google Scholar 

  330. Bo H, Kang W, Jiang N, Wang X, Zhang Y, Ji LL. Exercise-induced neuroprotection of hippocampus in APP/PS1 transgenic mice via upregulation of mitochondrial 8-oxoguanine DNA glycosylase. Oxid Med Cell Longev. 2014;2014:834502.

    Article  PubMed  PubMed Central  Google Scholar 

  331. Liang J, Wang C, Zhang H, Huang J, Xie J, Chen N. Exercise-induced benefits for Alzheimer’s disease by stimulating mitophagy and improving mitochondrial function. Front Aging Neurosci. 2021;13:755665.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  332. Podlesniy P, Figueiro-Silva J, Llado A, Antonell A, Sanchez-Valle R, Alcolea D, et al. Low cerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease. Ann Neurol. 2013;74:655–68.

    Article  CAS  PubMed  Google Scholar 

  333. Garcia-Mesa Y, Gimenez-Llort L, Lopez LC, Venegas C, Cristofol R, Escames G, et al. Melatonin plus physical exercise are highly neuroprotective in the 3xTg-AD mouse. Neurobiol Aging. 2012;33(1124):e1113–29.

    Google Scholar 

  334. Tanaka T, Nishimura A, Nishiyama K, Goto T, Numaga-Tomita T, Nishida M. Mitochondrial dynamics in exercise physiology. Pflugers Arch. 2020;472:137–53.

    Article  CAS  PubMed  Google Scholar 

  335. Kitaoka Y, Ogasawara R, Tamura Y, Fujita S, Hatta H. Effect of electrical stimulation-induced resistance exercise on mitochondrial fission and fusion proteins in rat skeletal muscle. Appl Physiol Nutr Metab. 2015;40:1137–42.

    Article  CAS  PubMed  Google Scholar 

  336. Li B, Liang F, Ding X, Yan Q, Zhao Y, Zhang X, et al. Interval and continuous exercise overcome memory deficits related to beta-Amyloid accumulation through modulating mitochondrial dynamics. Behav Brain Res. 2019;376:112171.

    Article  PubMed  CAS  Google Scholar 

  337. Koo JH, Kang EB, Cho JY. Resistance exercise improves mitochondrial quality control in a rat model of sporadic inclusion body myositis. Gerontology. 2019;65:240–52.

    Article  CAS  PubMed  Google Scholar 

  338. Chuang CS, Chang JC, Cheng FC, Liu KH, Su HL, Liu CS. Modulation of mitochondrial dynamics by treadmill training to improve gait and mitochondrial deficiency in a rat model of Parkinson’s disease. Life Sci. 2017;191:236–44.

    Article  CAS  PubMed  Google Scholar 

  339. Balan E, Schwalm C, Naslain D, Nielens H, Francaux M, Deldicque L. Regular endurance exercise promotes fission, mitophagy, and oxidative phosphorylation in human skeletal muscle independently of age. Front Physiol. 2019;10:1088.

    Article  PubMed  PubMed Central  Google Scholar 

  340. He W, Wang P, Chen Q, Li C. Exercise enhances mitochondrial fission and mitophagy to improve myopathy following critical limb ischemia in elderly mice via the PGC1a/FNDC5/irisin pathway. Skelet Muscle. 2020;10:25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  341. Landrigan JF, Bell T, Crowe M, Clay OJ, Mirman D. Lifting cognition: a meta-analysis of effects of resistance exercise on cognition. Psychol Res. 2020;84:1167–83.

    Article  PubMed  Google Scholar 

  342. Lamb SE, Sheehan B, Atherton N, Nichols V, Collins H, Mistry D, et al. Dementia And Physical Activity (DAPA) trial of moderate to high intensity exercise training for people with dementia: randomised controlled trial. BMJ. 2018;361:k1675.

    Article  PubMed  PubMed Central  Google Scholar 

  343. Baker LD, Frank LL, Foster-Schubert K, Green PS, Wilkinson CW, McTiernan A, et al. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch Neurol. 2010;67:71–9.

    Article  PubMed  PubMed Central  Google Scholar 

  344. Montagne A, Zhao Z, Zlokovic BV. Alzheimer’s disease: a matter of blood-brain barrier dysfunction? J Exp Med. 2017;214:3151–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  345. Benek O, Korabecny J, Soukup O. A Perspective on multi-target drugs for Alzheimer’s disease. Trends Pharmacol Sci. 2020;41:434–45.

    Article  CAS  PubMed  Google Scholar 

  346. Kwon TG, Park E, Kang C, Chang WH, Kim YH. The effects of combined repetitive transcranial magnetic stimulation and transcranial direct current stimulation on motor function in patients with stroke. Restor Neurol Neurosci. 2016;34:915–23.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the National Key Research and Development Program of China (2017YFB0403801), the National Natural Science Foundation of China (31971096, 31771256, and 32100918), the project funded by the China Postdoctoral Science Foundation (2021M690060), and the Sigma Xi Grants in Aid of Research (GIAR) program (G03152021115804390).

Author information

Authors and Affiliations

Authors

Contributions

CW, LMY, and LDY reviewed the literature and drafted the manuscript. CW and LMY drew the figures and tables. LDY, TCL, and RD supervised the writing and edited the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Luodan Yang, Timon Cheng-Yi Liu or Rui Duan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Supplementary Table 1.

Non-invasive Therapies in Clinical Trials.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, C., Yang, L., Feng, S. et al. Therapeutic non-invasive brain treatments in Alzheimer’s disease: recent advances and challenges. Inflamm Regener 42, 31 (2022). https://doi.org/10.1186/s41232-022-00216-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41232-022-00216-8

Keywords