Skip to main content

A review on traditional Chinese medicine natural products and acupuncture intervention for Alzheimer’s disease based on the neuroinflammatory

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease with insidious onset and progressive development. It is clinically characterized by cognitive impairment, memory impairment and behavioral change. Chinese herbal medicine and acupuncture are important components of traditional Chinese medicine (TCM), and are commonly used in clinical treatment of AD. This paper systematically summarizes the research progress of traditional Chinese medicine natural products and acupuncture treatment of AD, which combined with existing clinical and preclinical evidence, based on a comprehensive review of neuroinflammation, and discusses the efficacy and potential mechanisms of traditional Chinese medicine natural products and acupuncture treatment of AD. Resveratrol, curcumin, kaempferol and other Chinese herbal medicine components can significantly inhibit the neuroinflammation of AD in vivo and in vitro, and are candidates for the treatment of AD. Acupuncture can alleviate the memory and cognitive impairment of AD by improving neuroinflammation, synaptic plasticity, nerve cell apoptosis and reducing the production and aggregation of amyloid β protein (Aβ) in the brain. It has the characteristics of early, safe, effective and benign bidirectional adjustment. The purpose of this paper is to provide a basis for improving the clinical strategies of TCM for the treatment of AD.

Graphical Abstract

Introduction

Neurodegenerative disease is a kind of chronic and progressive nervous system disease, which is characterized by the uncontrolled loss of neurons and the accumulation of specific proteins in the brain and spinal cord, accompanied by the decline of brain function, especially cognitive function and motor function [1]. It includes the most common Alzheimer’s disease (AD), Parkinson’s disease (PD) characterized by static tremor, multiple sclerosis (MS) characterized by motor defects, and amyotrophic lateral sclerosis (ALS) characterized by progressive muscle weakness [2]. According to the latest data from the World Health Organization, the number of people living with dementia was around 55 million in 2019 and is expected to rise to 139 million by 2050, according to Word Alzheimer Report 2022. It seriously threatens people's life and health and brings heavy economic burden to society. Different neurodegenerative diseases have similar etiology at the cellular level, such as amyloid aggregation of proteins [3], neuroinflammation [1], oxidative stress [4], mitochondrial dysfunction, neuronal apoptosis [5], etc. However, they have their own unique symptoms. For example, AD is mainly manifested by sensory memory decline and progressive loss of cognitive function [6], while PD is mainly manifested by sensory motor loss [7].

AD is a neurodegenerative disease with occult onset and progressive development [8]. It is clinically characterized by cognitive impairment, memory impairment and behavioral change. Currently, the drugs approved by the U.S. Food and Drug Administration (FDA) for the treatment of AD are symptomatic treatments. These include acetylcholinesterase inhibitors (tacrine, donepezil, galantamine, and carbalatine) [9], the N-methyl-D-aspartate (NMDA) receptor antagonist memantine, and aducanumab, which targets Aβ [10]. Unfortunately, these symptomatic medications are often associated with side effects, including nausea, diarrhea, vomiting, and hypertensive crises. Therefore, developing a safe and effective new drug or therapy for treating AD is very important.

TCM is a traditional medicine originated from China, which has the characteristics of holistic concept and treatment based on syndrome differentiation. It mainly includes internal treatment based on Chinese herbal medicine and external treatment based on acupuncture. Since ancient times, more and more evidence has proved that Chinese herb medicine and acupuncture can effectively improve the clinical symptoms of AD [11,12,13]. AD belongs to the category of "dementia" in traditional Chinese medicine. The name of dementia first appeared in the Biography of the Divine Doctor Hua Tuo in the Han Dynasty. The key location of the disease lies in the brain and kidney, followed by the spleen, heart and liver. The disease can be divided into two categories: deficiency is caused by Yin deficiency and essence deficiency, brain and pulp insufficiency, and the solid is caused by phlegm turbidness, temperament and blood stasis. Tonifying kidney and essence is the basic principle of TCM treatment of AD, which runs through the whole treatment of AD [14]. The main mechanisms include reducing the production and aggregation of Aβ, inhibiting the phosphorylation of tau protein, inhibiting neuroinflammation, reducing oxidative stress and so on [15]. This paper first summarized the progress of the pathogenesis of AD, reviewed the clinical status of the treatment of AD by TCM, and systematically reviewed and discussed the progress of the therapeutic effect and pharmacological mechanism of effective components of traditional Chinese medicine natural products and acupuncture on AD based on neuroinflammation, in order to provide more reliable evidence for the treatment of AD by TCM.

Hypothesis of pathogenesis of AD

The main pathologic features of AD are extracellular Aβ plaque deposition and intracellular neurofibrillary tangles (NFTs). The pathogenesis of AD has not been fully elucidated [16]. At present, the pathogenesis of AD mainly consists of hypotheses such as amyloid and tau proteins, neuroinflammation, oxidative stress and cholinergic (Fig. 1).

Fig. 1
figure 1

An overview of pathogenesis of AD. (1) In the amyloid-producing pathway, Aβ is produced by the abnormal cleavage of APP by β-secretase 1 (BACE1) and γ-secretase. First, BACE1 cleaved the APP to produce sAPP and C-99 fragments. The C-99 fragment was cleaved by γ-secretase to produce two AICD fragments and an insoluble Aβ fragment. (2) Under normal circumstances, tau can bind to tubulin and stabilize microtubules, playing an important role in maintaining cell polarization, axonal transport and promoting neuronal growth. When tau is over-phosphorylated, tau dissociates from microtubules and aggregates into helical filaments (PHFS), which further aggregates to form NFTs, destabilizing microtubules and ultimately leading to neurofibrillary degeneration. (3) ACh is synthesized by acetyl-CoA and choline and catalyzed by choline acetyltransferase. Acetylcholinesterase in the synaptic cleft terminates signaling by hydrolyzing ACh. (4) Aβ stimulates the activation of microglia and astrocytes and induces reactive gliosis and proinflammatory signaling cascades. Activated microglia and astrocytes reduced Aβ clearance. (5) ROS promote the course of AD mainly through macromolecular peroxidation, Aβ metal ion REDOX potential and mitochondrial dysfunction, and affect cellular homeostasis, free radical production and increase the production of Aβ and p-tau

β-amyloid and tau hypothesis

The accumulation of Aβ protein and its accumulation and deposition in amyloid plaques are considered as the key pathogenesis of AD [17]. Aβ protein is a transmembrane protein that is produced by hydrolyzing Aβ precursor protein (APP). The first pathological change of AD is mainly the deposition of Aβ protein in the hippocampus [18]. The Aβ protein deposited in the hippocampus and basal segments in the form of amyloid plaques may recruit more Aβ protein to accumulate, thereby forming insoluble aggregates, resulting in mitochondrial damage, disrupting homeostasis, and synaptic dysfunction [19]. Microglia [20] and astrocytes [21, 22] are activated and induce the associated inflammatory response and OS, causing neuronal dysfunction and apoptosis, eventually leading to the development of AD [23, 24]. Tau protein kinase 1 is also activated by Aβ, resulting in abnormal phosphorylation of microtubule-associated proteins and promoting the formation of paired helical filaments (PHFs) and neurofibrillary tangles (NFTs), thereby accelerating the development of AD [25, 26].

Tau protein is a microtubule-associated protein produced by alternative splicing of MAPT gene [27]. The functions of tau protein include maintaining microtubule structure and assisting cytoplasmic transport, maintaining synaptic structure and function, and regulating neuronal signal transduction [28,29,30]. Tau protein is a phosphoprotein whose phosphorylation and dephosphorylation depend on the balance of protein kinase and protein phosphatase activity [31]. Normally, there are a limited number of tau phosphorylation sites, and they negatively regulate tau binding to microtubules. Under pathological conditions, tau phosphorylation sites may reach saturation, and the highly phosphorylated tau protein in the brain of AD patients may lead to its configuration change and loss of tubulin polymerization ability, resulting in impaired microtubule function [31, 32]. A high level of intracellular tau is involved in tau-tau interactions that may form insoluble PHFs and straight filaments (SFs), resulting in the formation of intracranial fibrous deposits, namely NFTs [33]. NFTs can reduce the number of synapses, produce neurotoxicity, and cause cellular dysfunction [34]. In addition, the acetylation and truncation of tau inhibit its ability to bind to microtubules, promote the occurrence of tau aggregation, and induce the occurrence of mitochondrial dysfunction and synaptic defects [35, 36].

Neuroinflammation hypothesis

Neuroinflammation is an inflammatory reaction in the CNS characterized by activation of glial cells [37]. Chronic inflammation is a key driver of neurodegenerative diseases, such as AD [38]. Neuroinflammation in AD is mainly driven by microglia (MG) [39, 40], which accounts for 5–10% of all glial cells in the brain, and it belongs to the mononuclear macrophage system. Oligodendrocyte function can maintain and protect the normal function of neurons. When oligodendrocyte is abnormal, it may cause demyelinating disease in the CNS and neuronal damage in severe cases [41]. In this process, astrocytes, oligodendrocytes, neurons, vascular endothelial cells, and peripheral immune cells all participate in the occurrence and progression of neuroinflammation [42, 43]. Neuroinflammation is manifested by the enhanced activation of microglia and astrocytes, increased release of pro-inflammatory cytokines in the brain, increased permeability of blood–brain barrier (BBB), and recruitment of peripheral immune cells into the CNS, eventually leading to neuronal dysfunction. In the case of AD pathology, Aβ stimulates activation of MG and astrocytes and induces reactive glia proliferation and pro-inflammatory signaling cascades. Activated MG and astrocytes reduce Aβ removal. Therefore, inhibiting the occurrence and development of inflammatory reaction is an important strategy for treating neurodegenerative diseases [44].

Oxidative stress hypothesis

As a key factor of neuronal damage in neurodegenerative diseases, OS is considered as a common contributing factor in a cascade of diseases. OS regulates MS disease within the CNS, and it plays a crucial role in maintaining the development and metabolism of the nervous system [45]. The substances produced by OS are pro-inflammatory substances and they can induce inflammatory responses, which are mainly mediated by OS and ROS. The abnormal aggregation of Aβ protein induces the production of free radicals, and activates ROS to cause oxidative damage and induce OS response. The excessive generation of ROS can result in damage to proteins, lipids, and DNA, leading to irreversible neuronal death [46]. The progression of AD may be mitigated or delayed by inhibiting or reducing OS-induced damage in patients with AD.

Cholinergic hypothesis

ACh is an important neurotransmitter in the cholinergic system, which is used to maintain the stability of learning and memory processes. AD is caused by the defect of neurotransmitters in the brain of AD patients, which leads to the damage of cholinergic neurons. The decrease of acetylcholinesterase (AChE) and acetylcholintransferase (ChAT) activity is the main reason for the decrease of acetylcholine (Ach) concentration and cholinergic activity, which leads to the decline of cognitive function in AD patients.

In conclusion, the aggregation of Aβ will accelerate the abnormal aggregation of tau protein and the production of reactive oxygen species (ROS). When Aβ accumulates outside the cell to form Aβ plaques, ROS production will be further accelerated, causing OS and resulting in membrane damage. At the same time, the neuroinflammatory response, MG cell activation, cytokine release and astroglial cell proliferation and other complex cascades are induced. In addition, Aβ plaques will destroy synaptic receptors and promote the release of glutamate into the synaptic cleve before synapses. When glutamate is released too much and accumulates in the synaptic cleve or spills over, it will cause the activation of extra-synaptic NMDAR, thus inducing excitatory toxicity.

Clinical evidence of TCM for AD

By searching Pubmed database, Cochrane, and Clinical trial databases, a total of 17 representative clinical trials related to acupuncture or chinese herbal medicine were screened, including 13 randomized controlled trials (RCTs), 3 before and after controlled trials, and 1 clinical observation. TCM injections or granules were used in 11 cases, acupuncture and moxibustion intervention was used in 3 cases, and acupuncture combined with chinese herbal medicine intervention was used in 3 cases. Based on the existing data, this paper summarized the clinical application status of traditional Chinese medicine and acupuncture in the treatment of AD.

Clinical status of Chinese medicine in the treatment of AD

At present, neuropsychological scale evaluation is the main method to evaluate the efficacy of dementia drugs, which mainly includes cognitive function, overall function of patients assessed by clinicians, quality of life and mental symptoms [47]. The clinical trial quantified the effect of medication on dementia using neuropsychological assessments at baseline and at the end of treatment [48]. In clinical practice, different neuropsychological measurement scales should be selected according to different evaluation purposes. Now it is widely used in the evaluation scale of cognitive efficacy, including the Mini Mental State Inventory (MMSE), the AD Cognitive Assessment (ADAS cog), and the Montreal cognitive assessment (MoCA) [49]. The concept of activity of daily living (ADL) should be included in the comprehensive functional evaluation, but because of its prominent role in the life of patients [50], the evaluation of ADL has become an essential part of the efficacy evaluation of dementia, which mainly uses the ADL evaluation scale, Very few studies used the caregiver burden questionnaire (CBQ) to evaluate the ability of daily living of patients [51]. A large number of clinical studies have shown that traditional Chinese medicine or integrated traditional Chinese and Western medicine treatment can significantly improve the clinical symptoms and laboratory indicators of Alzheimer's disease in different stages of onset. The chinese medicine used in the treatment of AD mainly has tonifying class, activating blood circulation and removing blood stasis class, phlegm class, mind-awakening class. Di-tan decoction (DTD) [52] is composed of Arisaema Cum Bile, Pinelliae Rhizoma, Aurantii Immaturus Fructus, Poria, Citri Reticulatae Pericarpium, Acori Tatarinowii Rhizoma, Ginseng Radix, Bambusae in Taeniam Caulis, Glycyrrhizae Radix, Zingiberis Recens Rhizoma and dextin. Clinical studies have shown that DTD treatment can increase the ADAS-Cog and C-MMSE scores of AD patients, indicating that it can improve the emotional or cognitive symptoms of AD patients and has a certain safety [52]. Laboratory studies have shown that the memory impairment of AD model mice was significantly reduced by DTD. In the brain tissue in mice treated with DTD, acetylcholine (Ach) and acetylcholine transferase (ChAT) were significantly increased, while acetylcholine esterase (AchE) was decreased. Likewise, Tiaoxin Recipe (TXR), Bushen Recipe (BSR) [53, 54] Jiannao Yizhi Formula [55], Huannao Yicong Formula [56], Fuzhisan [57], Shenfu injection, Shenmai Injectio [58] and other different TCM compounds can improve the scores of ADAS-Cog, CM-SS, MoCA, MMSE and so on.

Clinical effects of acupuncture on AD

AD is categorized within the realm of dementia in traditional Chinese medicine. According to the principles of traditional Chinese medicine, the kidneys govern bone health and generate vital essence, and it is the sufficiency of this kidney essence that safeguards memory. A deficiency in kidney essence is believed to be the root cause of memory loss. Consequently, Chinese medicine places great emphasis on the approach of “tonifying the kidney and essence” as a primary method for AD treatment [59, 60]. In the context of acupuncture, acupoint selection and technique play pivotal roles in determining treatment efficacy. Manual acupuncture (MA) and electro-acupuncture (EA) represent the two most commonly employed acupuncture methods in clinical practice. Beyond merely stimulating qi through acupuncture, practitioners employ techniques, such as lifting and twisting to manipulate specific acupoints adequately. This approach aims to tonify deficiencies and eliminate excess conditions. For instance, drawing from traditional Chinese medicine principles and clinical experience, TAN Yan [61] identified that acupoints distributed along the 14 meridians, with a concentration in the head and distal extremities, bear significant relevance to dementia treatment [62]. In the case of AD, 24 acupoints are situated across 11 meridians, encompassing points, such as Dumai (GV), Renmai (CV), Pangguangjing (BL), Ganjing (LR), Danjing (GB), Shenjing (KI), Weijing (ST), Pijing (SP), Xinjing (HT), Xinbaojing (PC), and Sanjiaojing (TE). The selection of acupoints for vascular dementia (VD) treatment follows a similar pattern to AD. For VD, the primary 19 acupoints are located along nine meridians, namely GV, CV, BL, LR, GB, KI, ST, SP, and PC. An essential acupoint in kidney tonification is BL23, as it corresponds to the Backshu acupoint of the kidney and plays a crucial role in replenishing kidney essence. Notably, acupuncture at Baihui (GV20) is known for its ability to invigorate the brain, open orifices, and calm the mind. It is widely employed in both research on brain diseases and clinical treatments using traditional Chinese medicine. The combined use of these two acupoints can effectively tonify the kidney, enhance essence, and improve cognitive functions. Contemporary studies have also corroborated that GV20 and BL23 are the most frequently utilized acupoints in research on AD model mechanisms [62]. In a study involving 20 AD patients, a treatment regimen utilizing kidney reinforcement and blood activation acupuncture methods over 12 weeks was employed. Specific acupoints, including Baihui (GV 20), Shenshu (BL 23), Xuehai (SP 10), and Geshu (BL 17), were selected. This treatment was found to enhance the cognitive abilities of AD patients, and its potential mechanism could be associated with the reduction of lipid peroxidation in the brains of AD patients [63]. A clinical study showed that for mild or moderate AD patients, Danzhong (RN 17), Zhongwan (RN 12), Qihai (RN 6), Zusanli (ST 36), Waiguan (SJ 5), and Xuehai (SP 10) were used as the basic acupuncture points, and other acupuncture points were utilized according to patients’ different conditions [64]. On the basis of the comprehensive assessment performed by ADAS-Cog and other scoring systems, acupuncture can improve cognitive function, while the improvement of daily living activities is limited. A study [65] demonstrated that for individuals experiencing early dementia symptoms, a combination of acupuncture at Baihui (GV 20), Sishencong (EX-HN 1), Dazhui (GV 14), Guanyuan (CV 4) points, and the administration of YizhiJiannao Granules yielded superior results compared to the Western medicine group. This was evidenced by improvement in ADAS-Cog and MMSE scores, as well as enhanced blood flow velocity in the middle cerebral artery (MCA), all achieved with fewer associated side effects. Peng [66] demonstrated that combination of acupuncture and moxibustion GV20, EX-HN1, GV14, CV4, and Yizhijiannao Granule significantly improved the effectiveness of the treatment of AD.

Olfactory three needle is a type of acupuncture method, in which the needle of bilateral Yingxiang (LI20) acupoint penetrates inward and upward to the starting point of nasolabial groove, and the third needle penetrates through the nasal root from the one-inch needle of Yintang (DU29). Neuroanatomical studies have revealed that DU29 is situated within the ophthalmic branch of the trigeminal nerve area, while LI20 resides in the upper branch of the trigeminal nerve’s distribution region. Notably, the ophthalmic branch of the trigeminal nerve, specifically the nasociliary nerve, covers the nasal mucosa, including the olfactory epithelium area. This results in an overlapping distribution pattern of both the olfactory and trigeminal nerves. In both clinical and fundamental research settings, the implementation of the Olfactory Three-Needle technique for the management of AD and mild cognitive impairment (MCI) has yielded promising outcomes [67, 68]. Drawing from years of clinical trial experience, Professor Liu Zhibin [69] has concluded that “ Xiusanzhen” can ameliorate cognitive dysfunction in AD patients by acting through the olfactory pathway. Additionally, it significantly enhances learning and memory functions. This effect is believed to be mediated by the reduction of serum endothelin (ET) level, an increase in calcitonin gene-related peptide (CGRP) content, and an overall enhancement of blood circulation in AD patients, thereby leading to improvement in cognitive function.

In summary, clinical research on acupuncture’s role in treating AD has advanced significantly. Acupuncture therapy adheres to well-defined principles within traditional Chinese medicine, concentrating on reinforcing kidney essence, nourishing blood, regulating qi, and awakening the brain. Both manual acupuncture and EA continue to hold essential roles, delivering definite therapeutic benefits. Acupuncture point selection primarily centers around acupoints along the governor vessel, heart channel, pericardial channel, and kidney channel, with clearly defined reinforcement and purgation methods. The parameters for EA, including intensity, amplitude, and frequency, are also well-established. However, it is imperative to note that while the efficacy of acupuncture in treating AD is well-documented, further in-depth research is required to elucidate the specific mechanisms of action underlying these therapeutic effects. This continued investigation will provide a more comprehensive understanding of the treatment’s potential.

In conclusion, the treatment of AD by traditional Chinese medicine is one of the important means to reduce the adverse reactions of western medicine and increase the curative effect [13]. The combination of modern medicine and traditional medicine in the treatment of AD has clinical advantages of increasing curative effect and reducing the toxic and side effects (Table 1). But more high-quality randomized controlled trials are needed to demonstrate the effectiveness of TCM in treating AD.

Table 1 Clinical efficacies of integrated TCM and WM for AD treatment

Potential mechanisms of traditional Chinese medicine natural products and acupuncture for AD

Potential mechanism of traditional Chinese medicine natural products modulating neuroinflammation in the treatment of AD

Neuroinflammation is the main pathological feature of AD patients. A large number of preclinical studies have found that TCM compounds or monomers can improve the pathological process of neurodegenerative diseases by inhibiting neuroinflammation [70]. Phenolic compounds, flavonoids and terpenoids are the main monomers of traditional Chinese medicine for the clinical treatment of AD. They can reduce neuroinflammation in AD model rats or mice by multi-level, multi-pathway and multi-target, and have the effect of delaying aging and preventing central nervous degenerative diseases. (Table 2).

Table 2 Potential mechanisms of TCM natural products for AD

Potential mechanisms of polyphenol compounds in the prevention and treatment of AD

In recent years, the role of polyphenols on human health has been paid more and more attention. The main source of polyphenols is vegetables and fruits, which have good antioxidant activity and can effectively scavenge free radicals [71]. As a natural polyphenol compound, resveratrol is easily absorbed by oral administration and excreted in urine and feces after metabolism. A large number of studies have shown that resveratrol [72] has neuroprotective properties, which can improve mitochondrial function, increase the clearance of toxic proteins by CNS, and ultimately improve the spatial learning and memory ability of AD model rats by inhibiting neuroinflammation and direct antioxidant stress [73]. MG and their membrane receptors are key players of neuroinflammation in AD [74]. TLR4/NF-κB/NLRP3 is an important signaling pathway regulating neuroinflammation in microglia [75]. Unactivated NF-κB [76] is usually bound to the inhibitory protein IκB in the cytoplasm. Under the stimulation of Aβ and inflammatory cells, proteasome degradation is accelerated and phosphorylation of IκB inhibitory protein is enhanced, leading to the release and nuclear translocation of NF-κB [77]. With the increase of NF-κB transcription, NLRP3 [78] inflammasome is activated as a multiprotein complex in the cytoplasm. After assembly, the proinflammatory caspase-1 precursor protein is activated into aspase-1, and the inflammatory cytokines IL-1β, IL-6, IL-18 [79] are secreted to induce the immune response of the body. The establishment of a chronic inflammatory environment in the brain can lead to neuronal injury and eventually lead to inflammatory cascade, which further aggravates the course of AD. In vitro experiments showed that in Aβ-induced AD cell model, resveratrol can inhibit TLR4/NF-κB [80]、TXNIP / TRX / NLRP3 signaling and transcription activators [81]. 3xTg-AD mice were created using Psen1 M146V mutation combined with APPSwe and tauP301L genes. It is the closest animal model to familial AD [82]. It has the main neuropathological features of SP and NFT, and the brain shows important pathological changes of AD, such as neuronal death and synaptic loss. In the 3xTG-AD animal model, resveratrol decreased the neuroinflammation and accumulation of Aβ oligomers, increased the levels of neurotrophin, synaptic markers, silencing information modulators, and decreased the markers of apoptosis, autophagy, endolysosomal degradation, and ubiquitination in the brain of 3xTG-AD mice [83]. Silent type information regulation 2 homolog1 (Sirt1) plays an important role in regulating cell stress, metabolism, growth, aging and apoptosis [84]. SIRT1 can reduce the content of Aβ by increasing the proportion of α-secretase of APP or decreasing BACE1 activity. In aluminum chloride (AlCl) and d-galactose (d-gal)—induced AD model mice, it was found that resveratrol can also increase the growth of neurites by activating SIRT1 and inhibiting the expression of microRNA-134 [85]. Through the regulation of these pathways, the overactivation of microglia can be inhibited to play a neuroprotective role.

Curcumin is a kind of yellow small molecule plant polyphenol extracted from the rhizome of turmeric, Curcuma, turmeric, etc. Numerous studies have provided experimental evidence for curcumin in the treatment and prevention of neurological diseases [86]. Curcumin can promote a series of neuroprotective responses by regulating various signal cascades in the brain, such as improving neuronal vitality, promoting neuronal differentiation and inhibiting neuronal apoptosis [87]. This involves a variety of mechanisms, including effects on neurotransmitters in the brain, modulation of the hypothalamic-pituitary-adrenocortical axis [88], upregulation of neurotrophic factor levels, or increased nerve regeneration. In ICV-STZ-induced AD rat model, curcumin can improve neuroinflammation and play a positive role in improving recognition memory [89]. In vivo neuroinflammation model, microglia were overactivated by LPS stimulation, curcumin enhanced AMPK activation in brain regions, and glial fibrillary acidic protein (GFAP), an astrocyte marker, was significantly reduced [90]. In the brain of AD patients, metals can induce amyloid aggregation and toxicity. Therefore, metal chelation can reduce the neurotoxicity caused by amyloid aggregation and oxidation. One Cu2+ or Fe2+ can bind at least two molecules of curcumin, suggesting another mechanism by which curcumin acts on AD [91]. Due to its binding function to REDOX metals, curcumin can also inhibit the inflammatory damage caused by the NF-κB [92] pathway caused by metals.

Potential mechanisms of flavonoid compounds in the prevention and treatment of AD

Plant-derived flavonoids have a wide range of anti-inflammatory and antioxidant activities and can effectively block toxic pathways associated with neurodegenerative disease pathology [93], including kaempferol, quercetin, baicalin and so on.

Oxidative stress is also a kind of important pathological mechanism, in the classic of AD induced by LPS model, contains galangal phenol drugs can significantly improve the activity of catalase, thereby inhibiting oxidative damage in cells, at the same time it also helps to improve alpha-synuclein pathogenic protein expression and tyrosine hydroxylase exception [94]. In spinal cord injury (SCI) model, gavage administration of kaempferol can improve the recovery of hindlimb motor function and spinal cord injury after SCI [95]. In addition, kaempferol administration decreased microglial activation and oxidative stress levels in the spinal cord. In vitro studies showed that kaempferol inhibited LPS-induced microglial activation in BV-2 cells [96]. BV2 cells pretreated with kaempferol reduced reactive oxygen species (ROS) production by inhibiting NADPH oxidase [97], followed by inhibiting phosphorylation of p38 MAPK and JNK, and subsequently inhibiting nuclear translocation of NF-κB p65 to inhibit proinflammatory factor expression [95]. Kaempferol can inhibit NF-κB, PI3K/Akt [94, 98], HMGB1/TLR4 [99]and other inflammatory pathways to play a neuroprotective role in LPS-induced neuroinflammation.

Quercetin is widely found in flowers, leaves, buds, seeds and fruits of many plants, mostly in the form of glycosides, such as rutin, quercetin, hyperin and so on. Quercetin can be obtained by acid hydrolysis. Many studies have reported that quercetin has therapeutic potential for brain diseases [100]. Quercetin treatment has been found to have anticonvulsant activity, which is correlated with brain concentration [101]. In addition, quercetin has been reported to promote antidepressant effects by inhibiting antioxidant effects [102]. What is more, quercetin treatment has been reported to attenuate hypothalamic–pituitary–adrenal (HPA) axis dysregulation in a mouse model of mild traumatic brain injury [102]. Importantly, oral quercetin decreased insoluble Aβ levels in the cortex of amyloid transgenic mouse models [103]. Macrophage polarization plays essential and diverse roles in most diseases. Homeostasis dysfunction in M1/M2 [104] macrophage polarization causes pathological conditions and inflammation. Neuroinflammation is characterized by microglial activation and the concomitant production of pro-inflammatory cytokines, leading to numerous neurodegenerative diseases and psychiatric disorders. In the study, we found that quercetin effectively inhibited the expression of lipocalin-2 in both macrophages and microglial cells stimulated by lipopolysaccharides (LPS). The production of nitric oxide (NO) and expression levels of the pro-inflammatory cytokines, inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2, were also attenuated by quercetin treatment [105]. The results also showed that quercetin significantly reduced the expression levels of the M1 markers, such as interleukin (IL)-6, TNF-α, and IL-1β, in the macrophages and microglia [106]. In addition, quercetin markedly reduced the production of various ROS in the microglia [107]. The microglial phagocytic ability induced by the LPS was also effectively reduced by the quercetin treatment. Importantly, the quercetin increased the expression levels of the M2 marker, IL-10, and the endogenous antioxidants, heme oxygenase (HO)-1, glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier subunit (GCLM), and NADPH quinone oxidoreductase-1 (NQO1) [108]. The enhancement of the M2 markers and endogenous antioxidants by quercetin was activated by the AMP-activated protein kinase (AMPK) and Akt signaling pathways. Together, the study reported that the quercetin inhibited the effects of M1 polarization, including neuroinflammatory responses, ROS production, and phagocytosis. Moreover, the quercetin enhanced the M2 macrophage polarization and endogenous antioxidant expression in both macrophages and microglia [104]. Naringenin also has potential effects on macrophages. Studies have shown that naringenin can cross the blood–brain barrier, reduce Aβ deposits and restore memory function in transgenic AD mice [109]. Treatment of primary cultured microglia with Aβ1-42 significantly promoted M2 microglia polarization and inhibited Aβ1-42-induced M1 microglia activation after naringenin administration. Microglia play a key role in brain Aβ clearance through Aβ-degrading enzymes after phagocytosis. After naringenin treatment, these Aβ-degrading enzymes were down-regulated in M1 microglia and up-regulated in M2 microglia. Thus, naringenin increased Aβ-degrading enzymes in M2 microglia, possibly leading to A decrease in Aβ plaques [110].

Toll-like receptors (TLRs) [111] are a class of pattern recognition receptors associated with neuroinflammation, among which TLR4 is a key factor in the regulation of immune response in the process of central nervous system infection and injury. TLR4 is widely distributed in brain microglia and other cells [112]. When TLR4 pathway is specifically recognized by LPS, it will further activate downstream NF-κB, leading to the release of proinflammatory factors, and then induce nerve cell death through apoptosis and other pathways [113]. Among them, Scutellaria baicalensis is often used in clinical treatment of a variety of acute infectious diseases, such as anti-inflammatory, antipyretic, anti-endotoxin, etc. Its baicalein can down-regulate TLR4 protein in microglial BV2 cell inflammatory model, and has anti-neuroinflammation effect [78]. In the LPS-induced inflammatory model of BV2 cells, the cells were significantly activated, and the secretion of proinflammatory factors IL-6, IL-1β and TNF-α increased [114]. Baicalein has obvious inhibitory effect on the inflammatory reaction. When BV2 cells in the resting state are stimulated by various factors, the NF-κB [115] heterodimer P65-P50 in the cytoplasm will be released into the nucleus and bind to the downstream target genes, thus initiating the transcriptional expression of target genes. The results of nuclear translocation of NF-κB P65 by immunofluorescence detection showed that, After LPS stimulation, the expression of NF-κB p65 protein in BV2 cytoplasm was significantly reduced, and most of the NF-κB p65 protein migrated to the nucleus. At the same time, the protein expressions of i-NOS and COX-2 were significantly down-regulated after administration of baicalein, indicating that baicalein inhibited LPS-induced inflammation in BV-2 cells. Administration of baicalein reversed MPTP-induced motor dysfunction, loss of dopaminergic neurons, and pro-inflammatory cytokine elevation. Baicalein also inhibited NLRP3 and caspase-1 activation and suppressed gasdermin D (GSDMD)-dependent pyroptosis. Additionally, baicalein inhibited the activation and proliferation of disease-associated proinflammatory microglia [116].

Potential mechanisms of terpenoid compounds in the prevention and treatment of AD

In recent years, natural drugs with few side effects and high safety have been widely used to treat a variety of diseases, including AD. Ginseng has a long history of medicinal use as it improves health and slows the aging process. Ginsenoside Rg1 in the treatment of AD include improvement in Aβ and Tau pathologies [117], regulation of synaptic function and intestinal microflora, and reduction of inflammation, oxidative stress, and apoptosis. The underlying mechanisms mainly involve the regulation of PKC, MAPK, PI3K/Akt, CDK5, GSK-3β, BDNF/TrkB, PKA/CREB, FGF2/Akt, p21WAF1/CIP1, NF-κB, NLRP1, TLR3, and TLR4 signaling pathways [117]. NADPH oxidase (NOXs) is the main enzyme responsible for excessive ROS production in many tissues. Studies have shown that NOX2 promotes aging-related neuronal oxidative stress damage and brain function loss, and the expression of NOX2 is significantly increased in long-term cultured hippocampal neurons. Ginsenoside Rg1, the main active component of ginseng, attenuates H2O2 -induced neuronal injury by inhibiting NADPH oxidase 2 (NOX2) and nucleotide binding oligomeric enzyme (NOD) -like receptor protein 1(NLRP1) inflammasome activation in hippocampal neurons in vitro [118]. Ginsenoside Rg1 can inhibit NoX2-mediated neuronal oxidative stress and neuroinflammation in APP/PS1 AD mice [119]. The NOD-like receptor family with three pyridine domains (NLRP3) is the most studied inflammasome sensor receptor protein. NLRP3, together with apoptosis-associated spot-like protein (ASC) and caspase-1 precursor protease, is composed of NLRP3 inflammasome [120]. In studies of AD patients and AD transgenic mice, it has also been found that inflammasome-derived ASC can bind to Aβ in the extracellular space of cells and promote Aβ aggregation, leading to the production of downstream inflammatory factors and promoting inflammatory responses. Some studies have shown that ginsenoside Rg1 has anti-inflammatory effects [121, 122]. Ginsenoside Rg1 can inhibit the production of downstream inflammatory factors and enhance the phagocytosis of Aβ in the neuroinflammatory response of AD.

Oleuropein (OLE), a non-toxic penoid glycoside compound, is the main component in olive leaves. Scholars administered olive leaf extract continuously for 3 months through 5xFAD mouse model. Oleoin was found to reduce neuroinflammation mainly by inhibiting NF-κB pathway and inhibiting the activation of NLRP3 inflammasome and RAGE/HMGB1 pathway [123]. In addition, oleuropein pretreatment of SH-SY5Y cells for 24 h could alleviate the cell death induced by Aβ42 and copper-Aβ42. In the transgenic mouse (APPswe/PS1dE9) model, treated mice (OLE) showed significantly reduced amyloid plaque deposition in the cortex and hippocampus compared with control mice [124].

The pathogenesis of AD is complex, including Aβ hypothesis, tau hypothesis, cholinergic hypothesis, inflammation hypothesis, OS hypothesis and cholinergic hypothesis. Current drug therapy for AD can only relieve the symptoms and has a single target, but cannot reverse the course of AD. Chinese herbal compounds, as widely available natural compounds, which have been shown to play a protective role against AD through multiple targets, such as inhibiting Aβ production and aggregation, reducing tau protein hyperphosphorylation and aggregation, regulating cholinergic system, inhibiting neuroinflammation, reducing OS, inducing autophagy and antagonizing NMDAR (Fig. 2). Although more and more studies have been conducted to reveal the mechanism of TCM monomer compounds in the prevention and treatment of AD, the application of the results of in vitro and animal studies to the clinic remains to be further studied.

Fig. 2
figure 2

Potential mechanism of traditional Chinese medicine natural products in the treatment of AD. Different types of traditional Chinese medicine monomer compounds play different roles in the process of AD. Epigallocatechin gallate (EGCG), Curcumin (CUR), Oleuropein (OLE), Resveratrol (RSV), Icariin (ICA), Ginsenoside Rg1(Rg1) and Naringenin(NAR) could inhibit the aggregation of Aβ, and resveratrol could prevent the phosphorylation of tau and reduce the level of p-tau. EGCG and RSV inhibited cholinesterase activity. kaempferol(KAF), RSV,CUR,QUE, NAR, Baicalein(BAL), Luteolin(LUT), Verbascoside(VER) inhibited neuroinflammation, Quercetin(QUE),RSV, EGCG reduce oxidative damage and other different pathways play a multi-level, multi-pathway, multi-target drug effect to prevent and control AD

Potential mechanism of acupuncture modulating neuroinflammation in the treatment of AD

The meridians network the whole body, and the meridians line in the meridians. Acupuncture adjusts the meridians and collaterals, and then adjusts the whole body's meridians [125]. The regulating effect of acupuncture and its meridian and viscera function is more obvious than other meridian. The biological basis of the correlation between meridians acupoints and viscera is mainly segmented interaction of nervous system [126]. Acupuncture plays an important role in the treatment of AD [127], which may improve the daily living ability of AD patients more safely and effectively than drugs, and can enhance the effect of drugs on improving cognitive function. Animal experiments have compared the efficacy of acupuncture at different points in treating animal models of AD, and evaluated the specific effects and neuropathologic mechanisms of acupoint therapy (Table 3, Fig. 3).

Table 3 Potential mechanisms of acupuncture ingredients for AD
Fig. 3
figure 3

Potential mechanism of acupuncture in the treatment of AD. Acupuncture stimulation of different acupoints can play a role by inhibition production and aggregation of Aβ fibers, inhibit tau hyperphosphorylation, modulate the neurotransmitter release, inhibiting neuroinflammation, reducing oxidative damage and so on

Acupuncture reduces neuroinflammation by inhibiting the activation of MG and astrocytes

Neuroinflammation is an immune response activated by glial cells in the CNS. It mainly occurs in response to stimuli, such as nerve injury, infection, and toxins, or in response to autoimmunity. Neuroinflammation is closely associated with the progression of neurodegenerative diseases, including AD, PD, ALS, and multiple sclerosis [37]. Recent studies have indicated that neuroinflammation has emerged as the third prominent pathological feature, following Aβ deposition and NFTs in AD. Inflammatory responses assume a pivotal role in both the onset and progression of AD [128, 129].

Studies have shown that Aβ can bind to receptors on the surface of microglia and astrocytes, trigger the release of inflammatory cytokines and chemokines, lead to the occurrence of chronic inflammation, mediate neuroinflammation and neurotoxicity, and impair cognitive function. Numerous studies have demonstrated that EA treatment can inhibit the activation of microglia and astrocytes in the hippocampus of AD patients, and reduce the deposition of Aβ [130]. The anti-inflammatory effect is related to the increase of the levels of anti-inflammatory cytokines (e.g., interleukin-2 (IL-2), IL-4, IL-10, IL-13, etc.) [23, 131] and the decrease of the levels of pro-inflammatory cytokines (e.g., tumor necrosis factor-α (TNF-α), IL-1β, and IL-6) [132, 133]. Acupuncture exhibited to significantly improve the working memory and synaptic plasticity of AD mice by inhibiting the phosphorylation of p38MAPK and the over-activation of MG cells in the hippocampus [67], as well as reduction of synaptic ultrastructural degradation in AD mice [134].

In the CNS, triggering receptor expressed on myeloid cells 2 (TREM2) is mainly expressed on the surface of microglia cells, mediating the proliferation, differentiation, survival, autophagy, and the expression levels of inflammatory factors in microglia cells, highlighting its crucial role in AD pathogenesis. Besides, Aβ was recently characterized as a ligand of TREM2, as it could directly bind TREM2 and activate TREM2 signaling pathway [135]. In vivo, TREM2 expression level is mainly upregulated under pathological conditions. For instance, increased expression level of TREM2 have been identified in AD patients [136] and in Aβ and tau pathological mouse models [137]. Overexpression of TREM2 is thought to be related to the recruitment of microglia to Aβ plaques [138]. As senescence accelerated mouse prone 8 (SAMP8) mice age, they exhibited accelerated cognitive decline, accompanied by pathological alterations in the CNS, including the cortex and hippocampus. These age-related changes align with the parameters monitored in the experiment. SAMP8 mice serve as valuable models for investigating the mechanisms underlying aging, cognitive function, and dysfunction, as well as for assessing potential anti-aging and cognitive enhancement interventions. Jiang et al.’s research revealed that EA could enhance TREM2 protein level in SAMP8 mice, thereby improving spatial learning and memory abilities, enhancing hippocampal neuronal morphology, and reducing the expression level of Aβ1-42 protein [139]. Thus, acupuncture may affect the pathological process of AD by regulating neuroinflammation in the brain through MG, AS, and TREM2.

Many studies have found that there is an interaction between neuroinflammation and autophagy in the pathogenesis of neurodegenerative diseases. LPS-induced neuroinflammation can cause autophagic damage through ATG gene imbalance. Autophagy, as a conservative catabolic process, can degrade defective proteins or organelles in lysosomes and recycle important components in eukaryotic cells. Under normal circumstances, it can have a protective effect, but abnormal autophagy may lead to cell death. Increasing evidence suggests that dysfunctional autophagy is an important trigger for AD [140]. Research indicates that autophagy is involved in the pathogenesis of AD, including Aβ metabolism, tau pathology, synaptic function, and mitochondrial dysfunction. Therefore, autophagy and neuroinflammation are expected to be used in the treatment of AD [141]. Chenglong Xie [142] used computer-aided drug screening technology to identify 18 small molecules that can act as mitochondrial autophagy inducers, increasing the survival and function of glutamate and acetylcholine neurons, eliminating amyloid-β and tau pathology, and improving the pathological symptoms of AD. Zheng Xiaoyan's [143] research has demonstrated that electroacupuncture can significantly improve cognitive dysfunction in 5xFAD transgenic mice expressing Aβ. Electroacupuncture achieves this by inhibiting the AKT-MAPK1-MTORC1 pathway in the prefrontal cortex and hippocampus, activating TFEB, and promoting the autophagic degradation of APP/Aβ. Lin Wenjia's [144] research found that the acupuncture points GV24 and bilateral GB13 can alleviate memory impairment related to Alzheimer's disease by promoting the autophagic clearance of Aβ and NLRP3 inflammasomes mediated by TFEB/TFE3.Although the relationship between mitochondrial autophagy and the pathogenesis of AD is still under investigation, acupuncture's regulation of mitochondrial autophagy-mediated clearance of dysfunctional mitochondria has shown potential for intervening in the treatment of AD.

Acupuncture reduces neuroinflammation and Aβ protein production by inhibiting inflammatory response

TLR4/NF-κB/NLRP3 signaling pathway

Inflammasome plays a key role in neuroinflammatory pathways, and it may be a target for AD therapy. Inflammasomes are an important component of innate immunity. They are multiprotein complexes, consisting of caspase, apoptosis-related spot-like proteins (ASCs), and intracytoplasmic pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), recruit and activate pro-inflammatory protease Caspase-1, and activated Caspase-1 cleaves IL-13 and IL-18 precursors to produce corresponding mature cytokines [145]. Five major inflammasome types have been found, including NLRP1, NLRP3, NLRC4, IPAF, and AIM2 inflammasomes. Jing Jiang et al. [146] studied the relationship between EA and the expression level of NLRP-3 inflammasome in the hippocampus of AD animal models, and they found that EA could inhibit the inflammatory response in the hippocampus of SAMP8 mice. In addition, the possible mechanism of EA is to reduce the expression levels of IL-1β and NLRP3 inflammasomes-related proteins. Other studies have also demonstrated that acupuncture can reregulate the expression levels and activities of inflammasomes, reduce inflammatory response in brain, and improve memory deficit and synaptic plasticity. Chuan He et al. [147] found that acupuncture pretreatment could inhibit the protein expression levels of NLRP3, Caspase-1, and IL-1β, and reduce the amount of activated MG in the hippocampus of rats. Ning Ding et al. [148] showed that MA could negatively regulate the NLRP3/Caspase-1 pathway in the hippocampus of AD mice. Kun Li et al. [149] reported that EA intervention in Baihui (GV 20) and Shenting (GV 24) inhibited the elevation of the expression levels of NLRP3, Caspase-1, IL-1β, and IL-18, improved memory deficits, and impaired synaptic plasticity. APP/PS1 double transgenic mice serve as valuable models for AD research, expressing mutant human presenilin DeltaE9 and a fusion gene of human mouse preamyloid protein APPswe under the regulation of mouse prion promoters. The DeltaE9 mutation, arising from the ninth exon’s deletion in the human presenilin gene is associated with early-onset AD. Dong-Mei Liao's investigation [150] has revealed that EA could effectively mitigate the overactivation of the TLR4/NF-κB/NLRP3 signaling pathway, thus ameliorating CNS inflammation and enhancing cognitive function in APP/PS1 mice. These findings suggest that acupuncture may exert its effects through modulation of the TLR4/NF-κB signaling pathway to suppress inflammasome activation. Furthermore, acupuncture has shown to reduce Aβ protein accumulation in the hippocampus by inhibiting inflammasome activation. Ting Zhang’s research [151] demonstrated that EA could reduce Aβ production in the hippocampal tissue of SAMP8 mice, mitigate neuronal apoptosis and damage, and inhibit NLRP1 inflammasome activation in these mice.

PI3K/Akt signaling pathway

Yuan Wang et al.’ research [152] revealed that Olfactory Three-Needle acupuncture significantly enhanced the PI3K/AKT signaling pathway, leading to the phosphorylation inactivation of GSK-3β in AD. This effect could improve synaptic plasticity and neuronal survival. In Xinyu Huang et al.’s study [153], EA intervention led to significant reductions in Aβ and total tau protein levels in the hippocampus, as well as body weight. These changes were associated with modulation of the PI3K/Akt signaling pathway. Yinshan Tang et al.’s findings [154] indicated that EA could reverse cognitive deficits in AD model mice (APP/PS1) by inhibiting the JNK signaling pathway and modulating apoptotic signals. Zhitao Hou et al. [155] demonstrated that 10 HZ EA effectively alleviated learning and memory impairment in 7-month-old SAMP8 mice. This intervention reduced pathological hippocampal damage, enhanced synaptic structure, improved synaptic transmission, and regulated the expression levels of proteins related to the cAMP/PKA/CREB signaling pathway. Yan-Jun Wu et al.’s results [156] showed that EA effectively regulated the expression levels of proteins related to the PI3K/GSK3α signaling pathway in the hippocampal tissue of AD mice, reducing the formation and deposition of senile plaques (SPs). Dong Weiguo et al.’s research [157, 158] indicated that EA could mitigate synaptic loss, increase the expression levels of SYN and PSD95, and inhibit AMPK activation and eEF2K activity. These effects could be associated with the inhibition of the AMPK/eEF2K/eEF2 signaling pathway. Jian-Qiao Fang et al. [159] reported the decreased expression levels of p-p38 MAPK protein and IL-1β mRNA in the frontal lobe and hippocampus of AD rats following EA intervention. Li et al. found that acupuncture at RN17, RN12, RN4, SP10, and ST36 points significantly improved the cognitive function of SAMP8 mice, reduced inflammation, and attenuated nuclear damage by downregulation of the PI3K/PDK1/nPKC/Rac 1 signaling pathway [160]. Studies have demonstrated that the low activity of glucose transporters and insufficient glucose intake in the brain tissues of AD patients and animals could lead to glucose metabolism disorders in the brain, and the binding of advanced glycosylation terminal products with receptors in neuroinflammatory plaques and NFTs could further induce OS and promote the pathological process of AD.

GSK-3β signaling pathway

Acupuncture reduces neuroinflammation by inhibiting the production of Aβ protein and phosphorylation of Tan protein. NFT is the main pathological feature of AD. The hyperphosphorylated tau protein is an important component of NFT and one of the main influential factors of AD. Inhibiting tau protein phosphorylation is a crucial method to prevent and treat AD. GSK-3β is a serine/threonine protein kinase, which is involved in the regulation of various intracellular signal transduction pathways, and it plays a major role in gene expression, apoptosis, and neuronal plasticity [161] [58]. GSK-3β has two isoforms, α and β, which share 98% identity in the catalytic region, while differ slightly in the N-terminus and C-terminus. The activity of GSK-3β is regulated by phosphorylation level, and the activation of GSK-3β can inhibit some transcription factors that promote neuronal survival, while its inactivation can promote neuronal survival and improve the stability of cell structure [58]. Activation of Akt promotes phosphorylation of GSK-3β in the CNS. At resting state, GSK-3β can downregulate β-catenin level and inhibit neuronal proliferation, differentiation, and migration. Moreover,, it is important to consider the expression levels of Bcl-2 and Bax [162], downstream proteins of Akt, as they can influence the expression level of Caspase-3. In AD pathophysiology, GSK-3β plays a pivotal role, impacting various disease aspects, including tau phosphorylation, Aβ production, memory, neurogenesis, synaptic function, and serving as a potential therapeutic target for AD. Chao Yu et al.’s research demonstrated that prophylactic EA at GV20 and BL23 acupoints improved synaptic and neuronal microtubule damage in D-Galactose-induced AD rats. The underlying mechanism was associated with the inhibition of GSK3β/mTOR pathway activity, resulting in the reduced tau phosphorylation and enhanced autophagy activity [163]. Anping Xu et al. [164] demonstrated that EA treatment significantly enhanced cognitive ability and hippocampal glucose uptake in APP/PS1 mice. The phosphorylation of tau protein is inhibited by inducing AKT and GSK3β phosphorylation. Therefore, the AKT/GSK3β signaling pathway may play an irreplaceable role in the regulation process. Yang et al. [165] studied the effects of acupuncture on cognitive function and the mechanism of treatment, and they found that EA at GV24 and bilateral GB13 points could reduce the levels of Aβ, p-tau (s396), and p-tau (s404) in the brain. Wang et al. [166] found that EA could reduce the overexpression of phosphorylated tau protein (Ser199, Ser202) in hippocampus of rats to improve cognitive function in AD rats.

Acupuncture reduces neuroinflammation by improving synaptic plasticity

Aβ deposition can lead to the onset of AD, and reducing the accumulation of Aβ in the brain can delay or alleviate AD symptoms. A large number of studies have shown that Aβ is a common pathway induced by various causes of AD, and it plays a noticeable role in the formation and development of AD. In an Aβ 1–40-induced AD rat model, acupuncture administered at GV24 and bilateral GB13 revealed notable changes in cerebral glucose metabolism (CGM) within the hypothalamus, thalamus, and brain stem, as indicated by positron emission tomography (PET). These changes suggest that acupuncture may enhance the learning and memory capabilities of AD rats [167]. Additionally, EA treatment led to a reduction in Aβ deposition mediated by MG, aligning with decreased amyloid precursor protein level [130]. Lin-Mei Wang et al. [168] showed that EA treatment at GV20 and BL23 effectively reduced inflammatory response and Aβ level in APP/PS1 mice. EA also enhanced the autophagic state, improved lymphatic system clearance ability [169], and reduce intracellular Aβ [170] to improve the learning and memory abilities of mice. Studies [171,172,173] have shown that EA could reduce the accumulation of Aβ in the hippocampus of APP/PS1 mice and promote neurogenesis, and the mechanism may be related to energy metabolism [174] and synaptic regulation [175]. The 5xFAD mouse model, generated by combining three human APP mutants with two PS1 mutations, exhibited early-onset amyloidosis, cognitive impairment, and neuronal loss. EA at GV20 and BL23 [176] enhanced hippocampal synaptic transmission, aiding in synaptic injury recovery. Furthermore, EA at GV20, Dazhui (DU 14), and BL23 improved synaptic ultrastructure, while acupuncture at RN17, RN12, RN6, ST36, and SP10 enhanced dendritic structure [177]. High-frequency EA (50 Hz) proved more effective than low-frequency (2 Hz) or medium-frequency (30 Hz) EA [176]. Mudan Cai [130] was used in 5xFAD mice, bilateral EA treatment at KI3 points significantly enhanced working memory and synaptic plasticity, reduced neuroinflammation, and mitigated synaptic ultrastructure degradation by bolstering synaptic functions. Furthermore, Xiaoyan Zheng's [143] research also shows that three-needle EA intervening at the GV24 and bilateral GB13 acupoints with EA, spatial learning and fear memory in 5xFAD mice were significantly improved. This treatment approach also resulted in the decreased levels of APP, its C-terminal fragment (CTFs), and Aβ deposits, and inhibited glial cell activation in the prefrontal cortex and hippocampus of 5xFAD. Yu et al. [176] and Yimin Jiang et al. [178] assessed the improvement effect of acupuncture at different frequencies on AD mice, and they found that high-frequency EA exhibited promising results, effectively inhibiting the activity of GSK-3β in the presence of Aβ1-42. This inhibition led to a reduction in learning and memory impairments induced by Aβ1-42 and provided protection against damage to synaptic ultrastructure.

Ran Ma et al. [179] investigated the effects of EA and MA on the learning and memory abilities, the ultrastructural changes of neurons, and the downregulation of CDK5 and tau proteins in the hippocampus of SAMP8 mice. Yang et al. [179] demonstrated that the expression levels of phosphorylated tau protein and tau mRNA in the hippocampus of the 3-month-old EA group decreased compared with that of the 9-month-old EA group, indicating that early EA intervention could more effectively improve the learning and memory abilities of SAMP8 mice and inhibit the phosphorylation of tau protein in the hippocampus.

Acupuncture reduces neuroinflammation by alleviating OS

The production of free radicals is an important factor causing OS, and OS plays a noticeable role in the early onset of AD. Excessive OS can not only cause neural cell death, but also lead to brain tissue damage. Therefore, inhibiting OS may be an important measure to prevent AD. Chang et al. [180] reported that after undergoing acupuncture treatment, the cognitive function of mice was improved, the number of neurons increased, the levels of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were elevated, and the levels of superoxide anion and protein carbonyl were reduced. Therefore, acupuncture may delay the brain aging of SAMP8 mice by alleviating OS. Wu et al. [181] found that EA treatment significantly restored total antioxidant capacity (T-AOC) and attenuated the abnormal increase in levels of ROS, MDA and 8-hydroxy-2-deoxyguanosine (8-OH-dG). It can effectively improve the hippocampal neuron damage and counteract the abnormal increase of NOX2 level in AD rats. Through precise needling of DU24 and GB13 acupoints, Cai et al. [130] demonstrated significant enhancements in behavior, a reduction in OS, the elevated level of the neurotransmitter acetylcholine, diminished apoptosis of hippocampal neurons, and a decrease in the expression levels of genes and proteins associated with apoptosis.

Acupuncture can increase the abundance of intestinal flora and inhibit neuroinflammatory response

Inflammatory response is closely associated with the imbalance of intestinal microbial community [182]. Traditionally, AD research primarily centered on the brain. However, in recent years, there was a shift towards investigating the role of GM in this context. Extensive studies on GM’s influence on the nervous system have revealed substantial disparities in GM composition and metabolite content between AD patients and the general population [183]. Moreover, interventions involving the reconstruction of intestinal flora have demonstrated the capability to reduce neuroinflammation in the brain and significantly enhance cognitive function in patients with AD [184,185,186]. These findings suggest that the microbial-gut-brain axis is an important pathologic pathway, influencing the occurrence and development of AD [187]. Yue Zhang et al. [12] evaluated the effects of MA on BBB dysfunction in APP/PS1 mice, and they demonstrated that MA could positively regulate intestinal flora and BBB dysfunction, and reduce the levels of TNF-α and IL-1β. Jing Jiang et al. [188] reported that EA could balance the number and composition of intestinal microbiota in SAMP8 mice and significantly reduce the expression levels of IL-1β, IL-6, and TNF-α in serum and hippocampal tissue. Therefore, EA can improve the abundance of intestinal flora and inhibit neuroinflammatory response.

Collectively, the acupuncture can not only inhibit production and aggregation of Aβ protein and phosphorylation of tau protein, but also can inhibit neuroinflammation, promote neurogenesis, regulate synaptic plasticity of the CNS [189], alleviate OS, improve disorders of intestinal flora, modulate brain glucose metabolism, and exert an effect of neuroprotection on the CNS cells.

Conclusions and perspectives

Neurodegenerative diseases are characterized by a progressive loss of neuronal structure and function and an overactive inflammatory response. An increasing number of studies have shown that the pathological mechanisms of neurodegenerative diseases are complex and poorly understood. The onset of AD is slow or insidious, mainly manifested by cognitive decline, psychiatric symptoms and behavioral disorders, and gradual decline in the ability of daily living. Divided into three stages according to the degree of cognitive and physical deterioration, including mild, moderate and severe dementia, Aβ [190, 191] plaque deposition usually leads to the accumulation of misfolded proteins and the production of reactive oxygen species, causing oxidative stress [192], resulting in membrane damage, mitochondrial dysfunction [193,194,195], and neurotrophic damage [196,197,198],At the same time, it can induce neuroinflammatory reaction [199], microglia activation, cytokine release and astrogliosis cascade reaction, thereby producing cytokines and triggering a series of inflammatory reactions, leading to synaptic dysfunction and inducing neurotoxic effects (Fig. 1). Due to the accurate diagnosis is difficult in preclinical AD, treat the patient began to appear the symptom of mild cognitive dysfunction period, disease has entered the early and mid-mostly, with Aβ again just at this time of treatment is to control the disease, such as so on single targets, such as Aβ, Tau protein failure, new drug research and development of resistance to the AD In addition to Aβ and Tau proteins, the CD33 and TREM2 genes involved in the regulation of glial cell function have been targeted to develop new anti-AD drugs. Although many drugs are in clinical trials, only a small fraction of these drugs have been successfully developed and approved for the treatment of neurodegenerative diseases [200, 201]. Although neuroinflammation may not be the trigger of neurodegenerative diseases, persistent inflammation can create a vicious cycle between neuronal lesions, leading to more neuronal death. Therefore, our treatment needs to focus on inhibiting microglial activation, proinflammatory cytokines and excessive production of oxidative stress.

According to TCM theory, the brain is the "house of the primitive God", which controls the life and spiritual activities of human beings, stores the essence and connects with the pulp, and is the place of life consciousness. Encephalopathy has been a serious threat to human health since ancient times. The multi-target strategy of TCM in the treatment of encephalopathy with "tonic deficiency", "tonic stasis" and "eliminate phlegm" as the treatment principle can provide new ideas for drug research and development of complex diseases such as AD. Modern pharmacological studies have found that many traditional Chinese medicine ingredients, such as tonifying, activating blood circulation and removing blood stasis, resolving phlegm, and awakening the mind, can inhibit inflammation in vivo and in vitro, and can be used as candidate drugs for the treatment of AD. It plays a role in inhibiting Aβ production and aggregation, reducing p-tau protein level, inhibiting neuroinflammation, alleviating oxidative damage and inhibiting cholinesterase activity. Through cell and animal models, a large number of researchers have found that traditional Chinese medicine compounds, monomer compounds or other traditional Chinese medicine therapies, such as acupuncture and moxibustion, have shown good efficacy, which can slow down the process of AD by regulating microglia function, inhibiting inflammation, improving oxidative stress and nutritional nerve. However, studies have shown that malabsorption, rapid metabolism and systemic elimination, inefficient drug delivery systems, and selective permeability across the BBB [202] are also serious problems, which largely limit the bioavailability and neuroprotective effects of TCM in neurodegenerative diseases. With the development of pharmaceutical technology, nanotechnology [203] has been used to increase the permeability of the BBB and improve the bioavailability of drugs. Compared with resveratrol alone, liposome resveratrol [204, 205] has more obvious antioxidant, free radical scavenging and ROS production reduction effect. Therefore, how to improve the absorption and stability of TCM, structure and formulation improvements, and more technologies and strategies for combination therapy are being developed, which provide more opportunities for TCM treatment of AD.

Acupuncture and moxibustion in the treatment of AD has the advantages of early, safe, effective [206] and benign bidirectional adjustment [207]. Acupuncture stimulation of different acupoints can play a role by inhibiting the production and aggregation of Aβ, preventing excessive phosphorylation and aggregation of tau protein, regulating the cholinergic system, inhibiting neuroinflammation, reducing oxidative damage and so on.In recent years, brain imaging technologies, such as functional magnetic resonance imaging and positron emission tomography, have been used to assess brain responses to acupuncture in a dynamic, visual, and objective way. These techniques are frequently used to explore neurological mechanisms of responses to acupuncture in AD and provide neuroimaging evidence as well as starting points to elucidate the possible mechanisms [208]. To further study the mechanism of acupuncture on AD, scholars combine it with acupuncture and moxibustion theory and put forward different acupuncture and moxibustion rules and acupoint selection prescriptions. MA and EA [62] therapy still occupy an important position, and the curative effect is certain. Body acupuncture acupoints are mainly Du meridian, heart meridian, pericardial meridian and kidney meridian, and the reinforcing and reducing technique is clear. The intensity, amplitude and frequency of electroacupuncture stimulation also have clear parameter range. Some scholars have proved that acupuncture combined with Chinese herbal medicine is more effective and safer in the treatment of diabetic neuropathy [209]. Therefore, the combination of acupuncture and medicine is expected to become a new treatment for AD, and the organic combination of basic and clinical research needs to be further strengthened, improve research level, perfect the unified diagnosis and curative effect evaluation standard, to gradually achieve objective research, standardization, standardization, for a variety of different stitch to strengthen clinical design scientific, rigor, at the same time, It is necessary to continue to strengthen the organic combination of clinical research and experimental research. In experimental research, it is necessary to analyze not only the influence of acupuncture and moxibustion on various experimental indicators, but also further study and analyze the influence of various experimental indicators on the clinical effect of acupuncture and moxibustion, so that experimental research can better serve the clinical practice. Finally, in order to further prove the efficacy and safety of TCM in the treatment of AD, more randomized controlled trials with high accuracy, clinical safety, rigorous design and large sample size should be carried out, and the mechanism of compatibility principle should be further explored.

All in all, the purpose of this review is to scientific and systematic evaluation of TCM in the role of AD, in combination with the existing clinical and preclinical evidence, a comprehensive discussion and shows the TCM treatment for AD curative effect and potential mechanism. In short, the application of modern Chinese medicine theory and modern scientific and technological means to deeply explore the pathogenesis of neurodegenerative diseases, and provide more reliable evidence for the treatment of AD with Chinese medicine.

Availability of data and materials

No data was used for the research described in the article.

Abbreviations

AD:

Alzheimer’s disease

ADL:

Activity of daily living

AS:

Astrocytes

ALS:

Amyotrophic lateral sclerosis

Aβ:

Beta-amyloid

APP:

Aβ precursor protein

Ach:

Acetylcholine

AChE:

Acetylcholinesterase

ADAS-Cog:

AD Cognitive Assessment

AlCl:

Aluminum chloride

AMPK:

AMP-activated protein kinase

ASCs:

Apoptosis-related spot-like proteins

BBB:

Blood–brain barrier

BSR:

Bushen Recipe

CBQ:

Caregiver burden questionnaire

CNS:

Central nervors system

CGM:

Cerebral glucose metabolism

CGRP:

Calcitonin gene-related peptide

ChAT:

Acetylcholintransferase

CTFs:

C-terminal fragment

DAMPs:

Damage-associated molecular patterns

DTD:

Di-tan decoction

d-gal:

D-galactose

ET:

Endothelin

EA:

Electro-acupuncture

FDA:

Food and Drug Administration

GCLC:

Glutamate-cysteine ligase catalytic

GCLM:

Glutamate-cysteine ligase modifier

GFAP:

Glial fibrillary acidic protein

GSH-Px:

Glutathione peroxidase

HO:

Heme oxygenase

HI:

Hippocampus

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

LPS:

Lipopolysaccharides

MA:

Manual acupuncture

MCA:

Middle cerebral artery

MCI:

Mild cognitive impairment

MDA:

Malondialdehyde

MG:

Microglia

MoCA:

Montreal cognitive assessment

MS:

Multiple sclerosis,

MMSE:

Mini Mental State Inventory

NMDA:

N-methyl-D-aspartate

NFTs:

Neurofibrillary tangles

NLRP3:

NOD-like receptor family with three pyridine domains

NQO1:

NADPH quinone oxidoreductase-1

NO:

Nitric oxide

OS:

Oxidative Stress

OLE:

Oleuropein

PD:

Parkinson's disease

PHFs:

Paired helical filaments

PRRs:

Pattern recognition receptors

PAMPs:

Pathogen-associated molecular patterns

PET:

Positron emission tomography

PFC:

Prefrontal cortex

PRRs:

Pattern recognition receptors

PAMPs:

Pathogen-associated molecular patterns

PET:

Positron emission tomography

ROS:

Reactive oxygen species

RCTs:

Randomized controlled trials

rs-fMRI:

Resting-state fMRI

SCI:

Spinal cord injury

SOD:

Superoxide dismutase

SHRs:

Spontaneously hypertensive rats

SFs:

Straight filaments

SPs:

Senile plaques

SAMP8:

Senescence accelerated mouse prone 8

Sirt1:

Silent type information regulation 2 homolog1

TCM:

Traditional Chinese medicine

TLRs:

Toll-like receptors

TREM2:

Triggering receptor expressed on myeloid cells 2

T-AOC:

Total antioxidant capacity

TNF-α:

Tumor necrosis factor-α

TXR:

Tiaoxin Recipe

VD:

Vascular dementia

WHO:

World Health Organization

8-OH-dG:

8-Hydroxy-2-deoxyguanosine

References

  1. Armstrong R. What causes neurodegenerative disease? Folia Neuropathol. 2020;58:93–112.

    Article  PubMed  Google Scholar 

  2. Dressman D, Elyaman W, Cells T. A growing universe of roles in neurodegenerative diseases. Neuroscientist. 2022;28:335–48.

    Article  CAS  PubMed  Google Scholar 

  3. Ross CA, Poirier MA. Protein aggregation and neurodegenerative disease. Nat Med. 2004;10(Suppl):S10–7.

    Article  PubMed  Google Scholar 

  4. Mandal PK, Roy RG, Samkaria A. Oxidative stress: glutathione and its potential to protect methionine-35 of abeta peptide from oxidation. ACS Omega. 2022;7:27052–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sengupta U, Kayed R. Amyloid beta, Tau, and alpha-Synuclein aggregates in the pathogenesis, prognosis, and therapeutics for neurodegenerative diseases. Prog Neurobiol. 2022;214: 102270.

    Article  CAS  PubMed  Google Scholar 

  6. Abuelezz NZ, Nasr FE, AbdulKader MA, Bassiouny AR, Zaky A. MicroRNAs as potential orchestrators of Alzheimer’s disease-related pathologies: insights on current status and future possibilities. Front Aging Neurosci. 2021;13: 743573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Martins-Pinge MC, de Jager L, de Campos BH, Bezerra LO, Turini PG, Pinge-Filho P. Nitric oxide involvement in cardiovascular dysfunctions of Parkinson disease. Front Pharmacol. 2022;13: 898797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Breijyeh Z, Karaman R. Comprehensive review on Alzheimer’s disease: causes and treatment. Molecules. 2020;25:5789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Moreira N, Lima J, Marchiori MF, Carvalho I, Sakamoto-Hojo ET. Neuroprotective effects of cholinesterase inhibitors: current scenario in therapies for Alzheimer’s disease and future perspectives. J Alzheimers Dis Rep. 2022;6:177–93.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Tagliapietra M. Aducanumab for the treatment of Alzheimer’s disease. Drugs Today. 2022;58:465–77.

    Article  Google Scholar 

  11. Wang XS, Li JJ, Wang YS, Yu CC, He C, Huang ZS, Jiang T, Hao Q, Kong LH. Acupuncture and related therapies for the cognitive function of Alzheimer’s disease: a network meta-analysis. Iran J Public Health. 2021;50:2411–26.

    PubMed  PubMed Central  Google Scholar 

  12. Zhang Y, Ding N, Hao X, Zhao J, Zhao Y, Li Y, Li Z. Manual acupuncture benignly regulates blood-brain barrier disruption and reduces lipopolysaccharide loading and systemic inflammation, possibly by adjusting the gut microbiota. Front Aging Neurosci. 2022;14:1018371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Deng C, Chen H, Meng Z, Meng S. Roles of traditional chinese medicine regulating neuroendocrinology on AD treatment. Front Endocrinol. 2022;13: 955618.

    Article  Google Scholar 

  14. Zhang Y, Zhang M. Neuroprotective effects of Morinda officinalis How: Anti-inflammatory and antioxidant roles in Alzheimer’s disease. Front Aging Neurosci. 2022;14:963041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Campos-Pena V, Pichardo-Rojas P, Sanchez-Barbosa T, Ortiz-Islas E, Rodriguez-Perez CE, Montes P, Ramos-Palacios G, Silva-Adaya D, Valencia-Quintana R, Cerna-Cortes JF, Toral-Rios D. Amyloid beta, lipid metabolism, basal cholinergic system, and therapeutics in Alzheimer’s disease. Int J Mol Sci. 2022;23:12092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Thal DR, Tome SO. The central role of tau in Alzheimer’s disease: from neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull. 2022;190:204–17.

    Article  CAS  PubMed  Google Scholar 

  17. Gallardo G, Holtzman DM. Amyloid-beta and Tau at the crossroads of Alzheimer’s disease. Adv Exp Med Biol. 2019;1184:187–203.

    Article  CAS  PubMed  Google Scholar 

  18. Pletnikova O, Kageyama Y, Rudow G, LaClair KD, Albert M, Crain BJ, Tian J, Fowler D, Troncoso JC. The spectrum of preclinical Alzheimer’s disease pathology and its modulation by ApoE genotype. Neurobiol Aging. 2018;71:72–80.

    Article  CAS  PubMed  Google Scholar 

  19. Mohandas E, Rajmohan V, Raghunath B. Neurobiology of Alzheimer’s disease. Indian J Psychiatry. 2009;51:55–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. J Cell Biol. 2018;217:459–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dhapola R, Hota SS, Sarma P, Bhattacharyya A, Medhi B, Reddy DH. Recent advances in molecular pathways and therapeutic implications targeting neuroinflammation for Alzheimer’s disease. Inflammopharmacology. 2021;29:1669–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xie L, Zhang N, Zhang Q, Li C, Sandhu AF, Iii GW, Lin S, Lv P, Liu Y, Wu Q, Yu S. Inflammatory factors and amyloid beta-induced microglial polarization promote inflammatory crosstalk with astrocytes. Aging. 2020;12:22538–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kaur D, Sharma V, Deshmukh R. Activation of microglia and astrocytes: a roadway to neuroinflammation and Alzheimer’s disease. Inflammopharmacology. 2019;27:663–77.

    Article  PubMed  Google Scholar 

  25. Mueed Z, Rai PK, Kamal MA, Poddar NK. Decoding the Inter-Relationship between sleep disorders and Alzheimer’s disease pathogenesis. CNS Neurol Disord Drug Targets. 2021;20:723–35.

    Article  CAS  PubMed  Google Scholar 

  26. Maccioni RB, Munoz JP, Barbeito L. The molecular bases of Alzheimer’s disease and other neurodegenerative disorders. Arch Med Res. 2001;32:367–81.

    Article  CAS  PubMed  Google Scholar 

  27. Ruiz-Gabarre D, Carnero-Espejo A, Avila J, Garcia-Escudero V. What’s in a Gene? The outstanding diversity of MAPT. Cells. 2022;11:840.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bloom GS. Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014;71:505–8.

    Article  PubMed  Google Scholar 

  29. Medeiros R, Baglietto-Vargas D, LaFerla FM. The role of tau in Alzheimer’s disease and related disorders. CNS Neurosci Ther. 2011;17:514–24.

    Article  CAS  PubMed  Google Scholar 

  30. El Mammeri N, Dregni AJ, Duan P, Wang HK, Hong M. Microtubule-binding core of the tau protein. Sci Adv. 2022;8:eabo4459.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Liang SY, Wang ZT, Tan L, Yu JT. Tau Toxicity in Neurodegeneration. Mol Neurobiol. 2022;59:3617–34.

    Article  CAS  PubMed  Google Scholar 

  32. Billingsley ML, Kincaid RL. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. Biochem J. 1997;323(Pt 3):577–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Al-Hilaly YK, Marshall KE, Lutter L, Biasetti L, Mengham K, Harrington CR, Xue WF, Wischik CM, Serpell LC. An additive-free model for Tau self-assembly. Methods Mol Biol. 2023;2551:163–88.

    Article  PubMed  Google Scholar 

  34. Sultana R, Butterfield DA. Alterations of some membrane transport proteins in Alzheimer’s disease: role of amyloid beta-peptide. Mol Biosyst. 2008;4:36–41.

    Article  CAS  PubMed  Google Scholar 

  35. Choi H, Kim HJ, Yang J, Chae S, Lee W, Chung S, Kim J, Choi H, Song H, Lee CK, Jun JH, Lee YJ, Lee K, Kim S, Sim HR, Choi YI, Ryu KH, Park JC, Lee D, Han SH, Hwang D, Kyung J, Mook-Jung I. Acetylation changes tau interactome to degrade tau in Alzheimer’s disease animal and organoid models. Aging Cell. 2020;19: e13081.

    Article  CAS  PubMed  Google Scholar 

  36. Shin MK, Vázquez-Rosa E, Koh Y, Dhar M, Chaubey K, Cintrón-Pérez CJ, Barker S, Miller E, Franke K, Noterman MF, Seth D. Reducing acetylated tau is neuroprotective in brain injury. Cell. 2021;184:2715-2732 e23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Singh D. Astrocytic and microglial cells as the modulators of neuroinflammation in Alzheimer’s disease. J Neuroinflammation. 2022;19:206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Boyd RJ, Avramopoulos D, Jantzie LL, McCallion AS. Neuroinflammation represents a common theme amongst genetic and environmental risk factors for Alzheimer and Parkinson diseases. J Neuroinflammation. 2022;19:223.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Cai Z, Hussain MD, Yan LJ. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci. 2014;124:307–21.

    Article  CAS  PubMed  Google Scholar 

  40. Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Rev Neurol. 2010;6:193–201.

    Article  PubMed  Google Scholar 

  41. Bacmeister CM, Barr HJ, McClain CR, Thornton MA, Nettles D, Welle CG, Hughes EG. Motor learning promotes remyelination via new and surviving oligodendrocytes. Nat Neurosci. 2020;23:819–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kabba JA, Xu Y, Christian H, Ruan W, Chenai K, Xiang Y, Zhang L, Saavedra JM, Pang T. Microglia: housekeeper of the central nervous system. Cell Mol Neurobiol. 2018;38:53–71.

    Article  CAS  PubMed  Google Scholar 

  43. Chen WW, Zhang X, Huang WJ. Role of neuroinflammation in neurodegenerative diseases (Review). Mol Med Rep. 2016;13:3391–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Heneka MT, O’Banion MK, Terwel D, Kummer MP. Neuroinflammatory processes in Alzheimer’s disease. J Neural Transm. 2010;117:919–47.

    Article  CAS  PubMed  Google Scholar 

  45. Cieslak M, Wojtczak A. Role of purinergic receptors in the Alzheimer’s disease. Purinergic Signal. 2018;14:331–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sbai O, Djelloul M, Auletta A, Ieraci A, Vascotto C, Perrone L. AGE-TXNIP axis drives inflammation in Alzheimer’s by targeting Abeta to mitochondria in microglia. Cell Death Dis. 2022;13:302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wei YC, Chen CK, Lin C, Chen PY, Hsu PC, Lin CP, Shyu YC, Huang WY. Normative data of mini-mental state examination, montreal cognitive assessment, and alzheimer’s disease assessment scale-cognitive subscale of community-dwelling older adults in Taiwan. Dement Geriatr Cogn Disord. 2022;51:365–76.

    Article  CAS  PubMed  Google Scholar 

  48. Traikapi A, Kalli I, Kyriakou A, Stylianou E, Symeou RT, Kardama A, Christou YP, Phylactou P, Konstantinou N. Episodic memory effects of gamma frequency precuneus transcranial magnetic stimulation in Alzheimer’s disease: a randomized multiple baseline study. J Neuropsychol. 2022. https://doi.org/10.1111/jnp.12299.

    Article  PubMed  Google Scholar 

  49. Gao Y, Zhao X, Huang J, Wang S, Chen X, Li M, Sun F, Wang G, Zhong Y. Abnormal regional homogeneity in right caudate as a potential neuroimaging biomarker for mild cognitive impairment: a resting-state fMRI study and support vector machine analysis. Front Aging Neurosci. 2022;14: 979183.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Dubbelman MA, Sanchez J, Schultz AP, Rentz DM, Amariglio RE, Sikkes SAM, Sperling RA, Johnson KA, Marshall GA. Everyday functioning and entorhinal and inferior temporal tau burden in cognitively normal older adults. J Prev Alzheimers Dis. 2022;9:801–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Shankle WR, Nielson KA, Cotman CW. Low-dose propranolol reduces aggression and agitation resembling that associated with orbitofrontal dysfunction in elderly demented patients. Alzheimer Dis Assoc Disord. 1995;9:233–7.

    Article  CAS  PubMed  Google Scholar 

  52. Chua KK, Wong A, Kwan PW, Song JX, Chen LL, Chan AL, Lu JH, Mok V, Li M. The efficacy and safety of the Chinese herbal medicine Di-Tan decoction for treating Alzheimer’s disease: protocol for a randomized controlled trial. Trials. 2015;16:199.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Zhang J, Yang C, Wei D, Li H, Leung EL, Deng Q, Liu Z, Fan XX, Zhang Z. Long-term efficacy of Chinese medicine Bushen capsule on cognition and brain activity in patients with amnestic mild cognitive impairment. Pharmacol Res. 2019;146: 104319.

    Article  PubMed  Google Scholar 

  54. Lin SM, Wang J, Zhou RQ. Clinical study on treatment of Alzheimer’s disease from the viewpoint of Xin and Shen. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2003;23:583–6.

    PubMed  Google Scholar 

  55. Wang HC, Liu NY, Zhang S, Yang Y, Wang ZY, Wei Y, Liu JG, Pei H, Li H. Clinical experience in treatment of Alzheimer’s disease with Jiannao Yizhi Formula () and Routine Western medicine. Chin J Integr Med. 2020;26:212–8.

    Article  CAS  PubMed  Google Scholar 

  56. Yang Y, Liu JP, Fang JY, Wang HC, Wei Y, Cao Y, Liu JG, Liu LT, Li H. Effect and safety of Huannao Yicong formula () in patients with mild-to-moderate alzheimer’s disease: a randomized, double-blinded, donepezil-controlled trial. Chin J Integr Med. 2019;25:574–81.

    Article  PubMed  Google Scholar 

  57. Bi M, Tong S, Zhang Z, Ma Q, Zhang S, Luo Z, Zhang Y, Li X, Wang D. Changes in cerebral glucose metabolism in patients with mild-to-moderate Alzheimer’s disease: a pilot study with the Chinese herbal medicine fuzhisan. Neurosci Lett. 2011;501:35–40.

    Article  CAS  PubMed  Google Scholar 

  58. Chen S, Yao X, Liang Y, Mei W, Liu X, Zhang C. Alzheimer’s disease treated with combined therapy based on nourishing marrow and reinforcing Qi. J Tradit Chin Med. 2015;35:255–9.

    Article  CAS  PubMed  Google Scholar 

  59. Li Z, Tong Q, Xu H, Hu L, Zhao R, Zhou F, Pan W, Zhou L. Therapeutic effects of TianDiJingWan on the abeta 25–35-induced Alzheimer’s disease model rats. Evid Based Complement Alternat Med. 2015;2015: 307350.

    PubMed  PubMed Central  Google Scholar 

  60. Zeng P, Liu YC, Wang XM, Ye CY, Sun YW, Su HF, Qiu SW, Li YN, Wang Y, Wang YC, Ma J, Li M, Tian Q. Targets and mechanisms of Alpinia oxyphylla Miquel fruits in treating neurodegenerative dementia. Front Aging Neurosci. 2022;14:1013891.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Yan T, Ya-Li Z, Jia-Ni Z, Wan-Zhu B. Acupuncture in neuroinflammation regulation may be related to the inhibition of M1 microglial activation involved in dementia. Progr Biochem Biophys. 2020;47:888–99.

    Google Scholar 

  62. Park S, Lee JH, Yang EJ. Effects of acupuncture on Alzheimer’s disease in animal-based research. Evid Based Complement Alternat Med. 2017;2017:6512520.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhu H, Dong KL, Wu Y, Zhang T, Li RM, Dai SS, Wang HL. Influence of acupuncture on isoprostane in patients with Alzheimer’s disease. Zhongguo Zhen Jiu. 2010;30:18–21.

    CAS  PubMed  Google Scholar 

  64. Jia Y, Zhang X, Yu J, Han J, Yu T, Shi J, Zhao L, Nie K. Acupuncture for patients with mild to moderate Alzheimer’s disease: a randomized controlled trial. BMC Complement Altern Med. 2017;17:556.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Han H, Li X, Jiang HN, Xu K, Wang Y. Effect of early acupuncture on cognitive function in patients with vascular dementia after cerebral infarction. Zhongguo Zhen Jiu. 2021;41:979–83.

    PubMed  Google Scholar 

  66. Peng XW, Dong KL. Clinical observation on acupuncture combined with Yizhi Jiannao granules for treatment of Alzheimer’s disease. Zhongguo Zhen Jiu. 2009;29:269–71.

    PubMed  Google Scholar 

  67. Wang Y, Wang Q, Ren B, Guo T, Qiang J, Cao H, Gao Y, Liu Z, Gao X, Zhu B. “Olfactory three-needle” enhances spatial learning and memory ability in SAMP8 mice. Behav Neurol. 2020;2020:2893289.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Liu Z, Niu W, Yang X, Wang Y. Effects of combined acupuncture and eugenol on learning-memory ability and antioxidation system of hippocampus in Alzheimer disease rats via olfactory system stimulation. J Tradit Chin Med. 2013;33:399–402.

    Article  PubMed  Google Scholar 

  69. Bingzhi Liu, Wenmin iu, Xianghang Yang, and X. Niu. 2009. Effect of “Xiusanzhen” on Serum ET and CGRP in Alzheimer’s Disease. J Shaanxi Coll Tradit Chin Med. 32 (2009) 16–17.

  70. Singh A, Agarwal S, Singh S. Age related neurodegenerative Alzheimer’s disease: usage of traditional herbs in therapeutics. Neurosci Lett. 2020;717: 134679.

    Article  CAS  PubMed  Google Scholar 

  71. Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BY, Chen Q, Yu CL, Wong VK, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary plant polyphenols as the potential drugs in neurodegenerative diseases: current evidence, advances, and opportunities. Oxid Med Cell Longev. 2022;2022:5288698.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Sawda C, Moussa C, Turner RS. Resveratrol for Alzheimer’s disease. Ann N Y Acad Sci. 2017;1403:142–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Grinan-Ferre C, Bellver-Sanchis A, Izquierdo V, Corpas R, Roig-Soriano J, Chillon M, Andres-Lacueva C, Somogyvari M, Soti C, Sanfeliu C, Pallas M. The pleiotropic neuroprotective effects of resveratrol in cognitive decline and Alzheimer’s disease pathology: From antioxidant to epigenetic therapy. Ageing Res Rev. 2021;67: 101271.

    Article  CAS  PubMed  Google Scholar 

  74. Calsolaro V, Edison P. Neuroinflammation in Alzheimer’s disease: current evidence and future directions. Alzheimers Dement. 2016;12:719–32.

    Article  PubMed  Google Scholar 

  75. Zhou X, Yuan L, Zhao X, Hou C, Ma W, Yu H, Xiao R. Genistein antagonizes inflammatory damage induced by beta-amyloid peptide in microglia through TLR4 and NF-kappaB. Nutrition. 2014;30:90–5.

    Article  PubMed  Google Scholar 

  76. Vargas AM, Rivera-Rodriguez DE, Martinez LR. Methamphetamine alters the TLR4 signaling pathway, NF-kappaB activation, and pro-inflammatory cytokine production in LPS-challenged NR-9460 microglia-like cells. Mol Immunol. 2020;121:159–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Choi YH. Catalpol attenuates lipopolysaccharide-induced inflammatory responses in BV2 microglia through inhibiting the TLR4-mediated NF-kappaB pathway. Gen Physiol Biophys. 2019;38:111–22.

    Article  CAS  PubMed  Google Scholar 

  78. Jin X, Liu MY, Zhang DF, Zhong X, Du K, Qian P, Yao WF, Gao H, Wei MJ. Baicalin mitigates cognitive impairment and protects neurons from microglia-mediated neuroinflammation via suppressing NLRP3 inflammasomes and TLR4/NF-kappaB signaling pathway. CNS Neurosci Ther. 2019;25:575–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Luo W, Han Y, Meng P, Yang Q, Zhao H, Ling J, Wang Y. Resatorvid relieves breast cancer complicated with depression by inactivating hippocampal microglia through TLR4/NF-kappaB/NLRP3 signaling pathway. Cancer Manag Res. 2020;12:13003–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Rahimifard M, Maqbool F, Moeini-Nodeh S, Niaz K, Abdollahi M, Braidy N, Nabavi SM, Nabavi SF. Targeting the TLR4 signaling pathway by polyphenols: A novel therapeutic strategy for neuroinflammation. Ageing Res Rev. 2017;36:11–9.

    Article  CAS  PubMed  Google Scholar 

  81. Feng L, Zhang L. Resveratrol suppresses abeta-induced microglial activation through the TXNIP/TRX/NLRP3 signaling pathway. DNA Cell Biol. 2019;38:874–9.

    Article  CAS  PubMed  Google Scholar 

  82. Gimenez-Llort L, Blazquez G, Canete T, Johansson B, Oddo S, Tobena A, LaFerla FM, Fernandez-Teruel A. Modeling behavioral and neuronal symptoms of Alzheimer’s disease in mice: a role for intraneuronal amyloid. Neurosci Biobehav Rev. 2007;31:125–47.

    Article  CAS  PubMed  Google Scholar 

  83. Broderick TL, Rasool S, Li R, Zhang Y, Anderson M, Al-Nakkash L, Plochocki JH, Geetha T, Babu JR. Neuroprotective effects of chronic resveratrol treatment and exercise training in the 3xTg-AD mouse model of Alzheimer’s disease. Int J Mol Sci. 2020;21:7337.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Qi J, Fu LY, Liu KL, Li RJ, Qiao JA, Yu XJ, Yu JY, Li Y, Feng ZP, Yi QY, Jia H, Gao HL, Tan H, Kang YM. Resveratrol in the hypothalamic paraventricular nucleus attenuates hypertension by regulation of ROS and neurotransmitters. Nutrients. 2022;14:4177.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Abozaid OAR, Sallam MW, El-Sonbaty S, Aziza S, Emad B, Ahmed ESA. Resveratrol-selenium nanoparticles alleviate neuroinflammation and neurotoxicity in a rat model of Alzheimer’s disease by regulating sirt1/miRNA-134/GSK3beta expression. Biol Trace Elem Res. 2022. https://doi.org/10.1007/s12011-021-03073-7.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Sabouni N, Marzouni HZ, Palizban S, Meidaninikjeh S, Kesharwani P, Jamialahmadi T, Sahebkar A. Role of curcumin and its nanoformulations in the treatment of neurological diseases through the effects on stem cells. J Drug Target. 2022;31:1–34.

    Google Scholar 

  87. Joshi P, Bisht A, Joshi S, Semwal D, Nema NK, Dwivedi J, Sharma S. Ameliorating potential of curcumin and its analogue in central nervous system disorders and related conditions: a review of molecular pathways. Phytother Res. 2022;36:3143–80.

    Article  CAS  PubMed  Google Scholar 

  88. Wang LY, Raskind MA, Wilkinson CW, Shofer JB, Sikkema C, Szot P, Quinn JF, Galasko DR, Peskind ER. Associations between CSF cortisol and CSF norepinephrine in cognitively normal controls and patients with amnestic MCI and AD dementia. Int J Geriatr Psychiatry. 2018;33:763–8.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Bassani TB, Turnes JM, Moura ELR, Bonato JM, Coppola-Segovia V, Zanata SM, Oliveira R, Vital M. Effects of curcumin on short-term spatial and recognition memory, adult neurogenesis and neuroinflammation in a streptozotocin-induced rat model of dementia of Alzheimer’s type. Behav Brain Res. 2017;335:41–54.

    Article  CAS  PubMed  Google Scholar 

  90. Qiao P, Ma J, Wang Y, Huang Z, Zou Q, Cai Z, Tang Y. Curcumin prevents neuroinflammation by inducing microglia to transform into the M2-phenotype via CaMKKbeta-dependent activation of the AMP-activated protein kinase signal pathway. Curr Alzheimer Res. 2020;17:735–52.

    Article  CAS  PubMed  Google Scholar 

  91. Mari M, Carrozza D, Malavasi G, Venturi E, Avino G, Capponi PC, Iori M, Rubagotti S, Belluti S, Asti M, Ferrari E. Curcumin-based beta-diketo ligands for Ga(3+): thermodynamic investigation of potential metal-based drugs. Pharmaceuticals. 2022;15:854.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sarawi WS, Alhusaini AM, Fadda LM, Alomar HA, Albaker AB, Aljrboa AS, Alotaibi AM, Hasan IH, Mahmoud AM. Curcumin and nano-curcumin mitigate copper neurotoxicity by modulating oxidative stress, inflammation, and Akt/GSK-3beta signaling. Molecules. 2021;26:5591.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wang F, Zhang S, Zhang J, Yuan F. Systematic review of ethnomedicine, phytochemistry, and pharmacology of Cyperi Rhizoma. Front Pharmacol. 2022;13: 965902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cai M, Zhuang W, Lv E, Liu Z, Wang Y, Zhang W, Fu W. Kaemperfol alleviates pyroptosis and microglia-mediated neuroinflammation in Parkinson’s disease via inhibiting p38MAPK/NF-kappaB signaling pathway. Neurochem Int. 2022;152: 105221.

    Article  CAS  PubMed  Google Scholar 

  95. Liu Z, Yao X, Sun B, Jiang W, Liao C, Dai X, Chen Y, Chen J, Ding R. Pretreatment with kaempferol attenuates microglia-mediate neuroinflammation by inhibiting MAPKs-NF-kappaB signaling pathway and pyroptosis after secondary spinal cord injury. Free Radic Biol Med. 2021;168:142–54.

    Article  CAS  PubMed  Google Scholar 

  96. Velagapudi R, Jamshaid F, Lepiarz I, Katola FO, Hemming K, Olajide OA. The tiliroside derivative, 3-O- [(E)-(2-oxo-4-(p-tolyl) but-3-en-1-yl] kaempferol produced inhibition of neuroinflammation and activation of AMPK and Nrf2/HO-1 pathways in BV-2 microglia. Int Immunopharmacol. 2019;77: 105951.

    Article  CAS  PubMed  Google Scholar 

  97. Yang CC, Hsiao LD, Wang CY, Lin WN, Shih YF, Chen YW, Cho RL, Tseng HC, Yang CM. HO-1 upregulation by kaempferol via ROS-dependent Nrf2-ARE Cascade attenuates lipopolysaccharide-mediated intercellular cell adhesion molecule-1 expression in human pulmonary alveolar epithelial cells. Antioxidants. 2022;11:782.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Cheng X, Yang YL, Yang H, Wang YH, Du GH. Kaempferol alleviates LPS-induced neuroinflammation and BBB dysfunction in mice via inhibiting HMGB1 release and down-regulating TLR4/MyD88 pathway. Int Immunopharmacol. 2018;56:29–35.

    Article  CAS  PubMed  Google Scholar 

  99. Yang YL, Cheng X, Li WH, Liu M, Wang YH, Du GH. Kaempferol attenuates LPS-induced striatum injury in mice involving anti-neuroinflammation, maintaining BBB integrity, and down-regulating the HMGB1/TLR4 pathway. Int J Mol Sci. 2019;20:491.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Alaqeel NK, AlSheikh MH, Al-Hariri MT. Quercetin nanoemulsion ameliorates neuronal dysfunction in experimental Alzheimer’s disease model. Antioxidants. 2022;11:1986.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Nieoczym D, Socala K, Raszewski G, Wlaz P. Effect of quercetin and rutin in some acute seizure models in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2014;54:50–8.

    Article  CAS  PubMed  Google Scholar 

  102. Holzmann I, da Silva LM, Correa da Silva JA, Steimbach VM, de Souza MM. Antidepressant-like effect of quercetin in bulbectomized mice and involvement of the antioxidant defenses, and the glutamatergic and oxidonitrergic pathways. Pharmacol Biochem Behav. 2015;136:55–63.

    Article  CAS  PubMed  Google Scholar 

  103. Zhang X, Hu J, Zhong L, Wang N, Yang L, Liu CC, Li H, Wang X, Zhou Y, Zhang Y, Xu H, Bu G, Zhuang J. Quercetin stabilizes apolipoprotein E and reduces brain Abeta levels in amyloid model mice. Neuropharmacology. 2016;108:179–92.

    Article  CAS  PubMed  Google Scholar 

  104. Tsai CF, Chen GW, Chen YC, Shen CK, Lu DY, Yang LY, Chen JH, Yeh WL. Regulatory effects of quercetin on M1/M2 macrophage polarization and oxidative/antioxidative balance. Nutrients. 2021;14:67.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Lee B, Yeom M, Shim I, Lee H, Hahm DH. Protective effects of quercetin on anxiety-like symptoms and neuroinflammation induced by lipopolysaccharide in rats. Evid Based Complement Alternat Med. 2020;2020:4892415.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Gong X, Huang Y, Ma Q, Jiang M, Zhan K, Zhao G. Quercetin alleviates lipopolysaccharide-induced cell damage and inflammation via regulation of the TLR4/NF-kappaB pathway in bovine intestinal epithelial cells. Curr Issues Mol Biol. 2022;44:5234–46.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Han X, Xu T, Fang Q, Zhang H, Yue L, Hu G, Sun L. Quercetin hinders microglial activation to alleviate neurotoxicity via the interplay between NLRP3 inflammasome and mitophagy. Redox Biol. 2021;44: 102010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhu W, Ge M, Li X, Wang J, Wang P, Tai T, Wang Y, Sun J, Shi G. Hyperoside Attenuates Zearalenone-induced spleen injury by suppressing oxidative stress and inhibiting apoptosis in mice. Int Immunopharmacol. 2022;102: 108408.

    Article  CAS  PubMed  Google Scholar 

  109. Yang W, Zhou K, Zhou Y, An Y, Hu T, Lu J, Huang S, Pei G. Naringin dihydrochalcone ameliorates cognitive deficits and neuropathology in APP/PS1 transgenic mice. Front Aging Neurosci. 2018;10:169.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Yang Z, Kuboyama T, Tohda C. Naringenin promotes microglial M2 polarization and Abeta degradation enzyme expression. Phytother Res. 2019;33:1114–21.

    Article  CAS  PubMed  Google Scholar 

  111. Ran Y, Qie S, Gao F, Ding Z, Yang S, Tian G, Liu Z, Xi J. Baicalein ameliorates ischemic brain damage through suppressing proinflammatory microglia polarization via inhibiting the TLR4/NF-kappaB and STAT1 pathway. Brain Res. 2021;1770: 147626.

    Article  CAS  PubMed  Google Scholar 

  112. Gurram PC, Manandhar S, Satarker S, Mudgal J, Arora D, Nampoothiri M. Dopaminergic signaling as a plausible modulator of astrocytic toll-like receptor 4: a crosstalk between neuroinflammation and cognition. CNS Neurol Disord Drug Targets. 2022;22:539.

    Google Scholar 

  113. Chen F, Wu X, Yang J, Yu X, Liu B, Yan Z. Hippocampal Galectin-3 knockdown alleviates lipopolysaccharide-induced neurotoxicity and cognitive deficits by inhibiting TLR4/NF-small ka, cyrillicB signaling in aged mice. Eur J Pharmacol. 2022;936: 175360.

    Article  CAS  PubMed  Google Scholar 

  114. Yan JJ, Du GH, Qin XM, Gao L. Baicalein attenuates the neuroinflammation in LPS-activated BV-2 microglial cells through suppression of pro-inflammatory cytokines, COX2/NF-kappaB expressions and regulation of metabolic abnormality. Int Immunopharmacol. 2020;79: 106092.

    Article  CAS  PubMed  Google Scholar 

  115. Jugait S, Areti A, Nellaiappan K, Narwani P, Saha P, Velayutham R, Kumar A. Neuroprotective effect of baicalein against oxaliplatin-induced peripheral neuropathy: impact on oxidative stress, neuro-inflammation and WNT/beta-catenin signaling. Mol Neurobiol. 2022;59:4334–50.

    Article  CAS  PubMed  Google Scholar 

  116. Rui W, Li S, Xiao H, Xiao M, Shi J. Baicalein Attenuates Neuroinflammation by Inhibiting NLRP3/caspase-1/GSDMD Pathway in MPTP Induced Mice Model of Parkinson’s Disease. Int J Neuropsychopharmacol. 2020. https://doi.org/10.1093/ijnp/pyaa060.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Wu JJ, Yang Y, Wan Y, Xia J, Xu JF, Zhang L, Liu D, Chen L, Tang F, Ao H, Peng C. New insights into the role and mechanisms of ginsenoside Rg1 in the management of Alzheimer’s disease. Biomed Pharmacother. 2022;152: 113207.

    Article  CAS  PubMed  Google Scholar 

  118. Xu TZ, Shen XY, Sun LL, Chen YL, Zhang BQ, Huang DK, Li WZ. Ginsenoside Rg1 protects against H2O2induced neuronal damage due to inhibition of the NLRP1 inflammasome signalling pathway in hippocampal neurons in vitro. Int J Mol Med. 2019;43:717–26.

    CAS  PubMed  Google Scholar 

  119. Zhang H, Su Y, Sun Z, Chen M, Han Y, Li Y, Dong X, Ding S, Fang Z, Li W, Li W. Ginsenoside Rg1 alleviates Abeta deposition by inhibiting NADPH oxidase 2 activation in APP/PS1 mice. J Ginseng Res. 2021;45:665–75.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Stancu IC, Cremers N, Vanrusselt H, Couturier J, Vanoosthuyse A, Kessels S, Lodder C, Brone B, Huaux F, Octave JN, Terwel D, Dewachter I. Aggregated Tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 2019;137:599–617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lyu TJ, Zhang ZX, Chen J, Liu ZJ. Ginsenoside Rg1 ameliorates apoptosis, senescence and oxidative stress in ox-LDL-induced vascular endothelial cells via the AMPK/SIRT3/p53 signaling pathway. Exp Ther Med. 2022;24:545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Li J, Yang C, Zhang S, Liu S, Zhao L, Luo H, Chen Y, Huang W. Ginsenoside Rg1 inhibits inflammatory responses via modulation of the nuclear factorkappaB pathway and inhibition of inflammasome activation in alcoholic hepatitis. Int J Mol Med. 2018;41:899–907.

    CAS  PubMed  Google Scholar 

  123. Abdallah IM, Al-Shami KM, Yang E, Wang J, Guillaume C, Kaddoumi A. Oleuropein-rich olive leaf extract attenuates neuroinflammation in the Alzheimer’s disease mouse model. ACS Chem Neurosci. 2022;13:1002–13.

    Article  CAS  PubMed  Google Scholar 

  124. Omar SH, Scott CJ, Hamlin AS, Obied HK. Olive biophenols reduces Alzheimer’s pathology in SH-SY5Y cells and APPswe mice. Int J Mol Sci. 2018;20:125.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Zhou W, Benharash P. Effects and mechanisms of acupuncture based on the principle of meridians. J Acupunct Meridian Stud. 2014;7:190–3.

    Article  PubMed  Google Scholar 

  126. Lee IS, Chae Y. Exploring acupuncture actions in the body and brain. J Acupunct Meridian Stud. 2022;15:157–62.

    Article  PubMed  Google Scholar 

  127. Wang H, Xie Y, Zhang Q, Xu N, Zhong H, Dong H, Liu L, Jiang T, Wang Q, Xiong L. Transcutaneous electric acupoint stimulation reduces intra-operative remifentanil consumption and alleviates postoperative side-effects in patients undergoing sinusotomy: a prospective, randomized, placebo-controlled trial. Br J Anaesth. 2014;112:1075–82.

    Article  CAS  PubMed  Google Scholar 

  128. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:157–72.

    Article  PubMed  Google Scholar 

  129. Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial cell-mediated neuroinflammation in Alzheimer’s disease. Int J Mol Sci. 2022;23:10572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Cai M, Lee JH, Yang EJ. Electroacupuncture attenuates cognition impairment via anti-neuroinflammation in an Alzheimer’s disease animal model. J Neuroinflammation. 2019;16:264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Jiang MC, Liang J, Zhang YJ, Wang JR, Hao JD, Wang MK, Xu J. Effects of acupuncture stimulation of bilateral “Hegu” (LI 4) and “Taichong” (LR 3) on learning-memory ability, hippocampal AP 42 expression and inflammatory cytokines in rats with Alzheimer’s disease. Zhen Ci Yan Jiu. 2016;41:113–8.

    PubMed  Google Scholar 

  132. Xie L, Liu Y, Zhang N, Li C, Sandhu AF, Williams G 3rd, Shen Y, Li H, Wu Q, Yu S. Electroacupuncture improves M2 microglia polarization and glia anti-inflammation of hippocampus in Alzheimer’s disease. Front Neurosci. 2021;15: 689629.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Ynag JY, Jiang J, Tian HL, Wang ZD, Ren JY, Liu H, Li ZG. Effect of electroacupuncture on learning-memory ability and expression of IL-1beta, IL-6 and TNF-alpha in hippocampus and spleen in mice with Alzheimer’s disease. Zhen Ci Yan Jiu. 2021;46:353–61.

    PubMed  Google Scholar 

  134. Tang SH, Du YJ, Tao YM, Tian Q, Kong LH. Effect of acupuncture on the ultrastructure of neurons and astrocytes in the hippocampal dentate gyrus in rats with Alzheimer’s disease induced by Abeta(1–42). Zhongguo Zhen Jiu. 2019;39:281–6.

    PubMed  Google Scholar 

  135. Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, Sun Y, Zhu B, Pina-Crespo JC, Zhang M, Zhang N, Chen X, Bu G, An Z, Huang TY, Xu H. TREM2 is a receptor for beta-amyloid that mediates microglial function. Neuron. 2018;97:1023–10317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Lue LF, Schmitz CT, Serrano G, Sue LI, Beach TG, Walker DG. TREM2 protein expression changes correlate with Alzheimer’s disease neurodegenerative pathologies in post-mortem temporal cortices. Brain Pathol. 2015;25:469–80.

    Article  CAS  PubMed  Google Scholar 

  137. Perez SE, Nadeem M, He B, Miguel JC, Malek-Ahmadi MH, Chen K, Mufson EJ. Neocortical and hippocampal TREM2 protein levels during the progression of Alzheimer’s disease. Neurobiol Aging. 2017;54:133–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Celarain N, Sanchez-Ruiz de Gordoa J, Zelaya MV, Roldan M, Larumbe R, Pulido L, Echavarri C, Mendioroz M. TREM2 upregulation correlates with 5-hydroxymethycytosine enrichment in Alzheimer’s disease hippocampus. Clin Epigenetics. 2016;8:37.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Jiang J, Liu H, Wang Z, Tian H, Wang S, Yang J, Li Z. Effects of electroacupuncture on DNA methylation of the TREM2 gene in senescence-accelerated mouse prone 8 mice. Acupunct Med. 2022;40:463–9.

    Article  PubMed  Google Scholar 

  140. Tao X, Zhang R, Wang L, Li X, Gong W. Luteolin and exercise combination therapy ameliorates amyloid-beta1-42 oligomers-induced cognitive impairment in AD mice by mediating neuroinflammation and autophagy. J Alzheimers Dis. 2023;92:195–208.

    Article  CAS  PubMed  Google Scholar 

  141. Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, Lautrup S, Hasan-Olive MM, Caponio D, Dan X, Rocktaschel P, Croteau DL, Akbari M, Greig NH, Fladby T, Nilsen H, Cader MZ, Mattson MP, Tavernarakis N, Bohr VA. Mitophagy inhibits amyloid-beta and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci. 2019;22:401–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Xie C, Zhuang XX, Niu Z, Ai R, Lautrup S, Zheng S, Jiang Y, Han R, Gupta TS, Cao S, Lagartos-Donate MJ, Cai CZ, Xie LM, Caponio D, Wang WW, Schmauck-Medina T, Zhang J, Wang HL, Lou G, Xiao X, Zheng W, Palikaras K, Yang G, Caldwell KA, Caldwell GA, Shen HM, Nilsen H, Lu JH, Fang EF. Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow. Nat Biomed Eng. 2022;6:76–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Zheng X, Lin W, Jiang Y, Lu K, Wei W, Huo Q, Cui S, Yang X, Li M, Xu N, Tang C, Song JX. Electroacupuncture ameliorates beta-amyloid pathology and cognitive impairment in Alzheimer disease via a novel mechanism involving activation of TFEB (transcription factor EB). Autophagy. 2021;17:3833–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Lin W, Li Z, Liang G, Zhou R, Zheng X, Tao R, Huo Q, Su C, Li M, Xu N, Tang C, Song JX. TNEA therapy promotes the autophagic degradation of NLRP3 inflammasome in a transgenic mouse model of Alzheimer’s disease via TFEB/TFE3 activation. J Neuroinflammation. 2023;20:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19:477–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Jiang J, Ding N, Wang K, Li Z. Electroacupuncture could influence the expression of IL-1beta and NLRP3 inflammasome in hippocampus of Alzheimer’s disease animal model. Evid Based Complement Alternat Med. 2018;2018:8296824.

    Article  PubMed  PubMed Central  Google Scholar 

  147. He C, Huang ZS, Chen HR, Yu CC, Wang XS, Jiang T, Kong LH. Effect of pretreatment of acupuncture on learning-memory ability and related protein of NLRP3 inflammasome in hippocampus in Alzheimer’s disease like rats. Zhongguo Zhen Jiu. 2020;40:1323–7.

    PubMed  Google Scholar 

  148. Ding N, Jiang J, Lu M, Hu J, Xu Y, Liu X, Li Z. Manual acupuncture suppresses the expression of proinflammatory proteins associated with the NLRP3 Inflammasome in the Hippocampus of SAMP8 Mice. Evid Based Complement Alternat Med. 2017;2017:3435891.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Li K, Shi G, Zhao Y, Chen Y, Gao J, Yao L, Zhao J, Li H, Xu Y, Chen Y. Electroacupuncture ameliorates neuroinflammation-mediated cognitive deficits through inhibition of NLRP3 in presenilin1/2 conditional double knockout mice. Neural Plast. 2021;2021:8814616.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Liao DM, Pang F, Zhou M, Li Y, Yang YH, Guo X, Tang CL. Effect of electroacupuncture on cognitive impairment in APP/PS1 mice based on TLR4/NF-kappaB/NLRP3 pathway. Zhen Ci Yan Jiu. 2022;47:565–72.

    PubMed  Google Scholar 

  151. Zhang T, Guan B, Tan S, Zhu H, Ren D, Li R, Xiao L. Bushen huoxue acupuncture inhibits NLRP1 inflammasome-mediated neuronal pyroptosis in SAMP8 mouse model of Alzheimer’s disease. Neuropsychiatr Dis Treat. 2021;17:339–46.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Wang Y, Zheng A, Yang H, Wang Q, Ren B, Guo T, Qiang J, Cao H, Gao YJ, Xu L, Li H, He L, Liu ZB. “Olfactory three-needle” acupuncture enhances synaptic function in Abeta(1–42)-induced Alzheimer’s disease via activating PI3K/AKT/GSK-3beta signaling pathway. J Integr Neurosci. 2021;20:55–65.

    Article  CAS  PubMed  Google Scholar 

  153. Huang X, Huang K, Li Z, Bai D, Hao Y, Wu Q, Yi W, Xu N, Pan Y, Zhang L. Electroacupuncture improves cognitive deficits and insulin resistance in an OLETF rat model of Al/D-gal induced aging model via the PI3K/Akt signaling pathway. Brain Res. 2020;1740: 146834.

    Article  CAS  PubMed  Google Scholar 

  154. Tang Y, Xu A, Shao S, Zhou Y, Xiong B, Li Z. Electroacupuncture ameliorates cognitive impairment by inhibiting the JNK signaling pathway in a mouse model of Alzheimer’s disease. Front Aging Neurosci. 2020;12:23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Hou Z, Yang X, Li Y, Chen J, Shang H. Electroacupuncture enhances neuroplasticity by regulating the orexin A-mediated cAMP/PKA/CREB signaling pathway in senescence-accelerated mouse prone 8 (SAMP8) mice. Oxid Med Cell Longev. 2022;2022:8694462.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Wu YJ, Wu KH, Jiang R, Wang YY, Luo M, Xia QQ, Tang CL, Zhu SJ, Sheng HJ. Effect of electroacupuncture on the expression of insulin signal pathway related proteins in hippocampus in mice with Alzheimer’s disease. Zhongguo Zhen Jiu. 2020;40:513–8.

    PubMed  Google Scholar 

  157. Dong W, Guo W, Zheng X, Wang F, Chen Y, Zhang W, Shi H. Electroacupuncture improves cognitive deficits associated with AMPK activation in SAMP8 mice. Metab Brain Dis. 2015;30:777–84.

    Article  CAS  PubMed  Google Scholar 

  158. Dong W, Yang W, Li F, Guo W, Qian C, Wang F, Li C, Lin L, Lin R. Electroacupuncture improves synaptic function in SAMP8 mice probably via inhibition of the AMPK/eEF2K/eEF2 signaling pathway. Evid Based Complement Alternat Med. 2019;2019:8260815.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Fang JQ, Zhu SX, Zhang Y, Wang F, Zhu QY. Effect of electroacupuncture on expression of phosphorylated P 38 MAPK and IL-1beta in frontal lobe and hippocampus in rats with Alzheimer’s disease. Zhen Ci Yan Jiu. 2013;38:35–9.

    CAS  PubMed  Google Scholar 

  160. Li G, Zeng L, Cheng H, Han J, Zhang X, Xie H. Acupuncture administration improves cognitive functions and alleviates inflammation and nuclear damage by regulating phosphatidylinositol 3 kinase (PI3K)/phosphoinositol-dependent kinase 1 (PDK1)/novel protein kinase C (nPKC)/Rac 1 signaling pathway in senescence-accelerated prone 8 (SAM-P8) mice. Med Sci Monit. 2019;25:4082–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 1998;12:3499–511.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Zhang SJ, Su SH, Gao JF. Electroacupuncture improves learning-memory ability possibly by suppressing apoptosis and down-regulating expression of apoptosis-related proteins in hippocampus and cerebral cortex in immature mice with Alzheimer’s disease. Zhen Ci Yan Jiu. 2020;45:611–6.

    PubMed  Google Scholar 

  163. Yu CC, Wang J, Ye SS, Gao S, Li J, Wang L, Jiang T, Wang XS, Li BC, Shu Q, Lu W, Du YJ, Kong LH. Preventive electroacupuncture ameliorates D-Galactose-induced Alzheimer’s Disease-like pathology and memory deficits probably via inhibition of GSK3beta/mTOR signaling pathway. Evid Based Complement Alternat Med. 2020;2020:1428752.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Xu A, Zeng Q, Tang Y, Wang X, Yuan X, Zhou Y, Li Z. Electroacupuncture protects cognition by regulating tau phosphorylation and glucose metabolism via the AKT/GSK3beta signaling pathway in Alzheimer’s disease Model Mice. Front Neurosci. 2020;14: 585476.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Yang Y, Hu S, Lin H, He J, Tang C. Electroacupuncture at GV24 and bilateral GB13 improves cognitive ability via influences the levels of Abeta, p-tau (s396) and p-tau (s404) in the hippocampus of Alzheimer’s disease model rats. NeuroReport. 2020;31:1072–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wang YY, Gao M, Qiu GP, Xu J, Wu YJ, Xu Y, Wang JR, Sheng HJ, Zhu SJ. Effect of electroacupuncture on the P35/P25-cyclin-dependent kinase 5-Tau pathway in hippocampus of rats with Alzheimer’s disease. Zhen Ci Yan Jiu. 2020;45:194–201.

    PubMed  Google Scholar 

  167. Cui S, Xu M, Huang J, Wang QM, Lai X, Nie B, Shan B, Luo X, Wong J, Tang C. Cerebral responses to acupuncture at GV24 and bilateral GB13 in rat models of Alzheimer’s disease. Behav Neurol. 2018;2018:8740284.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Wang LM, Zhao TT, Zhou HP, Zhou ZY, Huang S, Ling YL, Shao SJ. Effect of electroacupuncture on recognition memory and levels of Abeta, inflammatory factor proteins and aquaporin 4 in hippocampus of APP/PS1 double transgenic mice. Zhen Ci Yan Jiu. 2020;45:431–7.

    CAS  PubMed  Google Scholar 

  169. Liang PZ, Li L, Zhang YN, Shen Y, Zhang LL, Zhou J, Wang ZJ, Wang S, Yang S. Electroacupuncture improves clearance of amyloid-beta through the glymphatic system in the SAMP8 mouse model of Alzheimer’s disease. Neural Plast. 2021;2021:9960304.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Li F, Li LN, Wang X, Bai Y, Jiawula A, Bu QY, Gao TK, Xue WG. Effect of electroacupuncture stimulation of “Baihui” (GV 20) and “ Yongquan” (KI 1) on expression of hippocampal amyloid-beta and low density lipoprotein receptor-related protein-1 in APP/PS 1 transgenic mice. Zhen Ci Yan Jiu. 2015;40:55.

    PubMed  Google Scholar 

  171. Sun RQ, Wang ZD, Zhao J, Wang S, Liu YZ, Liu SY, Li ZG, Wang X. Improvement of electroacupuncture on APP/PS1 transgenic mice in behavioral probably due to reducing deposition of Abeta in hippocampus. Anat Rec. 2021;304:2521–30.

    Article  CAS  Google Scholar 

  172. Guo HD, Tian JX, Zhu J, Li L, Sun K, Shao SJ, Cui GH. Electroacupuncture suppressed neuronal apoptosis and improved cognitive impairment in the AD model rats possibly via downregulation of notch signaling pathway. Evid Based Complement Alternat Med. 2015;2015: 393569.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Li X, Guo F, Zhang Q, Huo T, Liu L, Wei H, Xiong L, Wang Q. Electroacupuncture decreases cognitive impairment and promotes neurogenesis in the APP/PS1 transgenic mice. BMC Complement Altern Med. 2014;14:37.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Liu W, Zhuo P, Li L, Jin H, Lin B, Zhang Y, Liang S, Wu J, Huang J, Wang Z, Lin R, Chen L, Tao J. Activation of brain glucose metabolism ameliorating cognitive impairment in APP/PS1 transgenic mice by electroacupuncture. Free Radic Biol Med. 2017;112:174–90.

    Article  CAS  PubMed  Google Scholar 

  175. Yang G, Pei YN, Shao SJ, Gao YS, Zhang SJ, Hu C, Feng SN, Xue WG. Effects of electroacupuncture at “Baihui” and “Yongquan” on the levels of synaptic plasticity related proteins postsynaptic density-95 and synaptophysin in hippocampus of APP/PS1 mice. Zhen Ci Yan Jiu. 2020;45:310–4.

    PubMed  Google Scholar 

  176. Yu CC, Wang Y, Shen F, Kong LH, Wang YW, Zhou H, Tang L. High-frequency (50 Hz) electroacupuncture ameliorates cognitive impairment in rats with amyloid beta 1–42-induced Alzheimer’s disease. Neural Regen Res. 2018;13:1833–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Kan BH, Yu JC, Zhao L, Zhao J, Li Z, Suo YR, Han JX. Acupuncture improves dendritic structure and spatial learning and memory ability of Alzheimer’s disease mice. Neural Regen Res. 2018;13:1390–5.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Jiang Y, Lin Y, Tan Y, Shen X, Liao M, Wang H, Lu N, Han F, Xu N, Tang C, Song J, Tao R. Electroacupuncture ameliorates cerebrovascular impairment in Alzheimer’s disease mice via melatonin signaling. CNS Neurosci Ther. 2022. https://doi.org/10.1111/cns.14027.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Ma R, Kong LH, Qi FJ, He RY, Zheng Q, Lu W, Fu XP. Effect of electroacupuncture on cyclin-dependent kinase 5 and Tau protein in hippocampus of SAMP8 mice. Zhen Ci Yan Jiu. 2020;45:529–34.

    PubMed  Google Scholar 

  180. Chang S, Guo X, Li G, Zhang X, Li J, Jia Y, Nie K. Acupuncture promotes expression of Hsp84/86 and delays brain ageing in SAMP8 mice. Acupunct Med. 2019;37:340–7.

    Article  PubMed  Google Scholar 

  181. Wu G, Li L, Li HM, Zeng Y, Wu WC. Electroacupuncture ameliorates spatial learning and memory impairment via attenuating NOX2-related oxidative stress in a rat model of Alzheimer’s disease induced by Abeta1–42. Cell Mol Biol. 2017;63:38–45.

    Article  CAS  PubMed  Google Scholar 

  182. Kesika P, Suganthy N, Sivamaruthi BS, Chaiyasut C. Role of gut-brain axis, gut microbial composition, and probiotic intervention in Alzheimer’s disease. Life Sci. 2021;264: 118627.

    Article  CAS  PubMed  Google Scholar 

  183. Leblhuber F, Ehrlich D, Steiner K, Geisler S, Fuchs D, Lanser L, Kurz K. The Immunopathogenesis of Alzheimer’s Disease Is Related to the Composition of Gut Microbiota. Nutrients. 2021;13:361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Minter MR, Zhang C, Leone V, Ringus DL, Zhang X, Oyler-Castrillo P, Musch MW, Liao F, Ward JF, Holtzman DM, Chang EB, Tanzi RE, Sisodia SS. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Sci Rep. 2016;6:30028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Kohler CA, Maes M, Slyepchenko A, Berk M, Solmi M, Lanctot KL, Carvalho AF. The gut-brain axis, including the microbiome, leaky gut and bacterial translocation: mechanisms and pathophysiological role in Alzheimer’s disease. Curr Pharm Des. 2016;22:6152–66.

    Article  CAS  PubMed  Google Scholar 

  186. Tse JKY. Gut microbiota, nitric oxide, and microglia as prerequisites for neurodegenerative disorders. ACS Chem Neurosci. 2017;8:1438–47.

    Article  CAS  PubMed  Google Scholar 

  187. Shabbir U, Arshad MS, Sameen A, Oh DH. Crosstalk between gut and brain in Alzheimer’s disease: the role of gut microbiota modulation strategies. Nutrients. 2021;13:690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Jiang J, Liu H, Wang Z, Tian H, Wang S, Yang J, Ren J. Electroacupuncture could balance the gut microbiota and improve the learning and memory abilities of Alzheimer’s disease animal model. PLoS ONE. 2021;16: e0259530.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wang Y, Zheng A, Yang H, Wang Q, Ren B, Guo T, Qiang J, Cao H, Gao YJ, Xu L, Li H, He L, Liu ZB. “Olfactory three-needle” acupuncture enhances synaptic function in Abeta1-42-induced Alzheimer’s disease via activating PI3K/AKT/GSK-3beta signaling pathway. J Integr Neurosci. 2021;20:55–65.

    Article  CAS  PubMed  Google Scholar 

  190. Peng C, Trojanowski JQ, Lee VM. Protein transmission in neurodegenerative disease. Nat Rev Neurol. 2020;16:199–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018;21:1332–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Ghemrawi R, Khair M. Endoplasmic reticulum stress and unfolded protein response in neurodegenerative diseases. Int J Mol Sci. 2020;21:6127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Johnson J, Mercado-Ayon E, Mercado-Ayon Y, Dong YN, Halawani S, Ngaba L, Lynch DR. Mitochondrial dysfunction in the development and progression of neurodegenerative diseases. Arch Biochem Biophys. 2021;702: 108698.

    Article  CAS  PubMed  Google Scholar 

  194. Wu Y, Chen M, Jiang J. Mitochondrial dysfunction in neurodegenerative diseases and drug targets via apoptotic signaling. Mitochondrion. 2019;49:35–45.

    Article  CAS  PubMed  Google Scholar 

  195. Palmer CS, Anderson AJ, Stojanovski D. Mitochondrial protein import dysfunction: mitochondrial disease, neurodegenerative disease and cancer. FEBS Lett. 2021;595:1107–31.

    Article  CAS  PubMed  Google Scholar 

  196. Pronin AV, Gogoleva IV, Torshin IY, Gromovа OA. Neurotrophic effects of lithium stimulate the reduction of ischemic and neurodegenerative brain damage. Zh Nevrol Psikhiatr Im S S Korsakova. 2016;116:99–108.

    Article  CAS  PubMed  Google Scholar 

  197. Uddin MS, Mamun AA, Rahman MM, Jeandet P, Alexiou A, Behl T, Sarwar MS, Sobarzo-Sanchez E, Ashraf GM, Sayed AA, Albadrani GM, Peluso I, Abdel-Daim MM. Natural products for neurodegeneration: regulating neurotrophic signals. Oxid Med Cell Longev. 2021;2021:8820406.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Morel L, Domingues O, Zimmer J, Michel T. Revisiting the role of neurotrophic factors in inflammation. Cells. 2020;9:865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Heneka MT, McManus RM, Latz E. Inflammasome signalling in brain function and neurodegenerative disease. Nat Rev Neurosci. 2018;19:610–21.

    Article  CAS  PubMed  Google Scholar 

  200. Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol. 2010;9:702–16.

    Article  CAS  PubMed  Google Scholar 

  201. Hung SY, Fu WM. Drug candidates in clinical trials for Alzheimer’s disease. J Biomed Sci. 2017;24:47.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Cai Z, Qiao PF, Wan CQ, Cai M, Zhou NK, Li Q. Role of blood-brain barrier in Alzheimer’s disease. J Alzheimers Dis. 2018;63:1223–34.

    Article  CAS  PubMed  Google Scholar 

  203. Xie J, Shen Z, Anraku Y, Kataoka K, Chen X. Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials. 2019;224: 119491.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Chimento A, De Amicis F, Sirianni R, Sinicropi MS, Puoci F, Casaburi I, Saturnino C, Pezzi V. Progress to improve oral bioavailability and beneficial effects of resveratrol. Int J Mol Sci. 2019;20:1381.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Wang Y, Xu H, Fu Q, Ma R, Xiang J. Protective effect of resveratrol derived from Polygonum cuspidatum and its liposomal form on nigral cells in parkinsonian rats. J Neurol Sci. 2011;304:29–34.

    Article  CAS  PubMed  Google Scholar 

  206. Teja Y, Helianthi DR, Nareswari I. The role of medical acupuncture therapy in Alzheimer’s disease. Med Acupunct. 2021;33:396–402.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Yin W, Lv G, Li C, Sun J. Acupuncture therapy for Alzheimer’s disease: the effectiveness and potential mechanisms. Anat Rec. 2021;304:2397–411.

    Article  Google Scholar 

  208. Yu CC, Ma CY, Wang H, Kong LH, Zhao Y, Shen F, Wu M. Effects of acupuncture on Alzheimer’s disease: evidence from neuroimaging studies. Chin J Integr Med. 2019;25:631–40.

    Article  PubMed  Google Scholar 

  209. Yu Z, Zhang W, Li B, Bao P, Wang F, Sun J, Song G, Yin L, Nan Z. Efficacy and safety of acupuncture combined with Chinese herbal medicine for diabetic nephropathy: a protocol for systematic review and meta-analysis. Medicine. 2021;100: e27087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Yang SQ, Fan DH, Yuan ZX, Su MY, Zhang ZN, Lin Y, Lin HG. Abdominal acupoint thread embedding therapy based on “brain-intestinal connection” for mild-to-moderate Alzheimer’s disease and its effects on serum levels of APP and Abeta1-42. Zhongguo Zhen Jiu. 2021;41:1231–5.

    PubMed  Google Scholar 

  211. Yu L, Lin SM, Zhou RQ, Tang WJ, Huang PX, Dong Y, Wang J, Yu ZH, Chen JL, Wei L, Xing SL, Cao HJ, Zhao HB. Chinese herbal medicine for patients with mild to moderate Alzheimer disease based on syndrome differentiation: a randomized controlled trial. Zhong Xi Yi Jie He Xue Bao. 2012;10:766–76.

    Article  PubMed  Google Scholar 

  212. Perry NSL, Menzies R, Hodgson F, Wedgewood P, Howes MR, Brooker HJ, Wesnes KA, Perry EK. A randomised double-blind placebo-controlled pilot trial of a combined extract of sage, rosemary and melissa, traditional herbal medicines, on the enhancement of memory in normal healthy subjects, including influence of age. Phytomedicine. 2018;39:42–8.

    Article  CAS  PubMed  Google Scholar 

  213. Liu P, Kong M, Liu S, Chen G, Wang P. Effect of reinforcing kidney-essence, removing phlegm, and promoting mental therapy on treating Alzheimer’s disease. J Tradit Chin Med. 2013;33:449–54.

    Article  PubMed  Google Scholar 

  214. Liu NY, Pei H, Liu MX, Liu LT, Fu CG, Li H, Chen KJ. Efficacy and safety of Guilingji capsules () for treating mild-to-moderate cognitive impairment: study protocol for a randomized, double-blind, positive-controlled, multicenter and noninferiority trial. Chin J Integr Med. 2020;26:577–82.

    Article  CAS  PubMed  Google Scholar 

  215. Shi J, Wei M, Ni J, Sun F, Sun L, Wang J, Yu T, Wang K, Lv P, Wang Y, Zhang Y, Gao X, Gao X, Luo B, Mao S, Zhang B, Ren X, Yu F, Hu W, Yin P, Wu N, Liu X, Bi Q, Wang Y, Tian J, C.S. Group. Tianzhi granule improves cognition and BPSD of vascular dementia: a randomized controlled trial. J Transl Med. 2020;18:76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Labban S, Alghamdi BS, Alshehri FS, Kurdi M. Effects of melatonin and resveratrol on recognition memory and passive avoidance performance in a mouse model of Alzheimer’s disease. Behav Brain Res. 2021;402: 113100.

    Article  CAS  PubMed  Google Scholar 

  217. Dong X, Tang Y, Zhan C, Wei G. Green tea extract EGCG plays a dual role in Abeta42 protofibril disruption and membrane protection: a molecular dynamic study. Chem Phys Lipids. 2021;234: 105024.

    Article  CAS  PubMed  Google Scholar 

  218. Bao J, Liu W, Zhou HY, Gui YR, Yang YH, Wu MJ, Xiao YF, Shang JT, Long GF, Shu XJ. Epigallocatechin-3-gallate alleviates cognitive deficits in APP/PS1 mice. Curr Med Sci. 2020;40:18–27.

    Article  CAS  PubMed  Google Scholar 

  219. Nan S, Wang P, Zhang Y, Fan J. Epigallocatechin-3-gallate provides protection against Alzheimer’s disease-induced learning and memory impairments in rats. Drug Des Devel Ther. 2021;15:2013–24.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Fan H, Tang HB, Shan LQ, Liu SC, Huang DG, Chen X, Chen Z, Yang M, Yin XH, Yang H, Hao DJ. Quercetin prevents necroptosis of oligodendrocytes by inhibiting macrophages/microglia polarization to M1 phenotype after spinal cord injury in rats. J Neuroinflammation. 2019;16:206.

    Article  PubMed  PubMed Central  Google Scholar 

  221. Alam Q, Krishnamurthy S. Dihydroquercetin ameliorates LPS-induced neuroinflammation and memory deficit. Curr Res Pharmacol Drug Discov. 2022;3: 100091.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Kou JJ, Shi JZ, He YY, Hao JJ, Zhang HY, Luo DM, Song JK, Yan Y, Xie XM, Du GH, Pang XB. Luteolin alleviates cognitive impairment in Alzheimer’s disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol Sin. 2022;43:840–9.

    Article  CAS  PubMed  Google Scholar 

  223. Feng J, Wang JX, Du YH, Liu Y, Zhang W, Chen JF, Liu YJ, Zheng M, Wang KJ, He GQ. Dihydromyricetin inhibits microglial activation and neuroinflammation by suppressing NLRP3 inflammasome activation in APP/PS1 transgenic mice. CNS Neurosci Ther. 2018;24:1207–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Zhu T, Zhang F, Li H, He Y, Zhang G, Huang N, Guo M, Li X. Long-term icariin treatment ameliorates cognitive deficits via CD4(+) T cell-mediated immuno-inflammatory responses in APP/PS1 mice. Clin Interv Aging. 2019;14:817–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Chen Y, Han S, Huang X, Ni J, He X. The protective effect of icariin on mitochondrial transport and distribution in primary hippocampal neurons from 3x Tg-AD mice. Int J Mol Sci. 2016;17:163.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Quan Q, Li X, Feng J, Hou J, Li M, Zhang B. Ginsenoside Rg1 reduces βamyloid levels by inhibiting CDΚ5induced PPAR&gamma, phosphorylation in a neuron model of Alzheimer’s disease. Mol Med Rep. 2020;22:3277–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Zhang Y, Ding S, Chen Y, Sun Z, Zhang J, Han Y, Dong X, Fang Z, Li W. Ginsenoside Rg1 alleviates lipopolysaccharide-induced neuronal damage by inhibiting NLRP1 inflammasomes in HT22 cells. Exp Ther Med. 2021;22:782.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Nie L, Xia J, Li H, Zhang Z, Yang Y, Huang X, He Z, Liu J, Yang X. Ginsenoside Rg1 ameliorates behavioral abnormalities and modulates the hippocampal proteomic change in triple transgenic mice of Alzheimer’s disease. Oxid Med Cell Longev. 2017;2017:6473506.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Zhang L, Xia R, Jia J, Wang L, Li K, Li Y, Zhang J. Oleanolic acid protects against cognitive decline and neuroinflammation-mediated neurotoxicity by blocking secretory phospholipase A2 IIA-activated calcium signals. Mol Immunol. 2018;99:95–103.

    Article  CAS  PubMed  Google Scholar 

  230. Chen S, Liu H, Wang S, Jiang H, Gao L, Wang L, Teng L, Wang C, Wang D. The neuroprotection of verbascoside in Alzheimer’s disease mediated through mitigation of neuroinflammation via blocking NF-kappaB-p65 signaling. Nutrients. 2022;14:1417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Jiang J, Liu G, Shi S, Li Y, Li Z. Effects of manual acupuncture combined with donepezil in a mouse model of Alzheimer’s disease. Acupunct Med. 2019;37:64–71.

    Article  PubMed  Google Scholar 

  232. Zhao L, Zhou C, Li L, Liu J, Shi H, Kan B, Li Z, Li Y, Han J, Yu J. Acupuncture improves cerebral microenvironment in mice with Alzheimer’s disease treated with hippocampal neural stem cells. Mol Neurobiol. 2017;54:5120–30.

    Article  CAS  PubMed  Google Scholar 

  233. Lai X, Ren J, Lu Y, Cui S, Chen J, Huang Y, Tang C, Shan B, Nie B. Effects of acupuncture at HT7 on glucose metabolism in a rat model of Alzheimer’s disease: an 18F-FDG-PET study. Acupunct Med. 2016;34:215–22.

    Article  PubMed  Google Scholar 

  234. Yang B, He M, Chen X, Sun M, Pan T, Xu X, Zhang X, Gong Q, Zhao Y, Jin Z, Cheng Z. Acupuncture effect assessment in APP/PS1 transgenic mice: on regulating learning-memory abilities, gut microbiota, and microbial metabolites. Comput Math Methods Med. 2022;2022:1527159.

    PubMed  PubMed Central  Google Scholar 

  235. Luo B, Zhao L, Zhang X, Kan B, Liu Y, Jia Y, Han J, Yu J. Acupuncture upregulates G protein coupled activity in SAMP8 mice. Acupunct Med. 2017;35:289–96.

    Article  PubMed  Google Scholar 

  236. Zhang J, Tang C, Liao W, Zhu M, Liu M, Sun N. The antiapoptotic and antioxidative stress effects of Zhisanzhen in the Alzheimer’s disease model rat. NeuroReport. 2019;30:628–36.

    Article  CAS  PubMed  Google Scholar 

  237. Yang JW, Wang XR, Ma SM, Yang NN, Li QQ, Liu CZ. Acupuncture attenuates cognitive impairment, oxidative stress and NF-kappaB activation in cerebral multi-infarct rats. Acupunct Med. 2019;37:283–91.

    Article  PubMed  Google Scholar 

  238. Li G, Zhang X, Cheng H, Shang X, Xie H, Zhang X, Yu J, Han J. Acupuncture improves cognitive deficits and increases neuron density of the hippocampus in middle-aged SAMP8 mice. Acupunct Med. 2012;30:339–45.

    Article  PubMed  Google Scholar 

  239. Zhao L, Jia Y, Yan D, Zhou C, Han J, Yu J. Aging-related changes of triose phosphate isomerase in hippocampus of senescence accelerated mouse and the intervention of acupuncture. Neurosci Lett. 2013;542:59–64.

    Article  CAS  PubMed  Google Scholar 

  240. Zhang M, Xv GH, Wang WX, Meng DJ, Ji Y. Electroacupuncture improves cognitive deficits and activates PPAR-gamma in a rat model of Alzheimer’s disease. Acupunct Med. 2017;35:44–51.

    Article  CAS  PubMed  Google Scholar 

  241. Lu Y, Huang Y, Tang C, Shan B, Cui S, Yang J, Chen J, Lin R, Xiao H, Qu S, Lai X. Brain areas involved in the acupuncture treatment of AD model rats: a PET study. BMC Complement Altern Med. 2014;14:178.

    Article  PubMed  PubMed Central  Google Scholar 

  242. Xu A, Tang Y, Zeng Q, Wang X, Tian H, Zhou Y, Li Z. Electroacupuncture enhances cognition by promoting brain glucose metabolism and inhibiting inflammation in the APP/PS1 mouse model of Alzheimer’s disease: a pilot study. J Alzheimers Dis. 2020;77:387–400.

    Article  CAS  PubMed  Google Scholar 

  243. Li Y, Jiang J, Tang Q, Tian H, Wang S, Wang Z, Liu H, Yang J, Ren J. Microglia TREM2: a potential role in the mechanism of action of electroacupuncture in an Alzheimer’s disease animal model. Neural Plast. 2020;2020:8867547.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Guo HD, Zhu J, Tian JX, Shao SJ, Xu YW, Mou FF, Han XJ, Yu ZH, Chen JL, Zhang DY, Zhang LS, Cui GH. Electroacupuncture improves memory and protects neurons by regulation of the autophagy pathway in a rat model of Alzheimer’s disease. Acupunct Med. 2016;34:449–56.

    Article  PubMed  Google Scholar 

  245. Li L, Li L, Zhang J, Huang S, Liu W, Wang Z, Liang S, Tao J, Chen L. Disease stage-associated alterations in learning and memory through the electroacupuncture modulation of the cortical microglial M1/M2 polarization in mice with Alzheimer’s disease. Neural Plast. 2020;2020:8836173.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Li L, Li J, Dai Y, Yang M, Liang S, Wang Z, Liu W, Chen L, Tao J. Electro-acupuncture improve the early pattern separation in Alzheimer’s disease mice via basal forebrain-hippocampus cholinergic neural circuit. Front Aging Neurosci. 2021;13: 770948.

    Article  CAS  PubMed  Google Scholar 

  247. Yang Q, Zhu S, Xu J, Tang C, Wu K, Wu Y, Wang Y, Sheng H. Effect of the electro-acupuncture on senile plaques and its formation in APP(+)/PS1(+) double transgenic mice. Genes Dis. 2019;6:282–9.

    Article  CAS  PubMed  Google Scholar 

  248. Ding N, Jiang J, Xu A, Tang Y, Li Z. Manual acupuncture regulates behavior and cerebral blood flow in the SAMP8 mouse model of Alzheimer’s disease. Front Neurosci. 2019;13:37.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work was supported by the National Natural Science Foundation of China (NSFC) (Nos. 81873369, 82030125, 82105024), Tianjin Science and Technology Project(No. 20JCQNJC00280), and the Youth Project of Tianjin Health Commission (No. 2021056) for financial support. We would like to thank Editage (www.editage.cn) for English language editing.

Funding

The work was supported by the National Natural Science Foundation of China (NSFC) (Nos. 81873369, 82030125, 82105024), Tianjin Graduate Research Innovation Project (2022BKY179) and TUTCM Graduate Research Innovation Project (YJSKC-20221020) for financial support.

Author information

Authors and Affiliations

Authors

Contributions

YG, XL and ZC conceptualized and designed the study, ZC and XW formed the literature analysis, and wrote the original manuscript draft. XL, ZX, QL and SD revised, edited and extended the final draft. All authors have reviewed and approved the manuscript before submission. All authors have reviewed and approved the manuscript before submission.

Corresponding authors

Correspondence to Yi Guo or Xiaowei Lin.

Ethics declarations

Ethics approval and consent to participate

Informed consent was obtained from all individual participants included in the study.

Consent for publication

All participants agree to publish.

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher's Note

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

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Z., Wang, X., Du, S. et al. A review on traditional Chinese medicine natural products and acupuncture intervention for Alzheimer’s disease based on the neuroinflammatory. Chin Med 19, 35 (2024). https://doi.org/10.1186/s13020-024-00900-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-024-00900-6

Keywords