Abstract
The global trend toward aging populations has resulted in an increase in the occurrence of Alzheimer's disease (AD) and associated socioeconomic burdens. Abnormal metabolism of amyloid‐β (Aβ) has been proposed as a significant pathomechanism in AD, supported by results of recent clinical trials using anti‐Aβ antibodies. Nonetheless, the cognitive benefits of the current treatments are limited. The etiology of AD is multifactorial, encompassing Aβ and tau accumulation, neuroinflammation, demyelination, vascular dysfunction, and comorbidities, which collectively lead to widespread neurodegeneration in the brain and cognitive impairment. Hence, solely removing Aβ from the brain may be insufficient to combat neurodegeneration and preserve cognition. To attain effective treatment for AD, it is necessary to (1) conduct extensive research on various mechanisms that cause neurodegeneration, including advances in neuroimaging techniques for earlier detection and a more precise characterization of molecular events at scales ranging from cellular to the full system level; (2) identify neuroprotective intervention targets against different neurodegeneration mechanisms; and (3) discover novel and optimal combinations of neuroprotective intervention strategies to maintain cognitive function in AD patients. The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline.
Keywords: Alzheimer's disease, early intervention, neural regeneration, neuroprotection, systematic perspective
The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline. The main neuroprotective intervention targets for AD including NTFs, antineuroinflammation, astrocytic protection, remyelination, stem‐cell therapy and in vivo neural regeneration, cerebral glucose metabolism regulation, vascular system protection, lymphatic protection, prion‐like proteinopathy regulation, peripheral neurotoxic substances clearance, systemic NTFs, and physical exercise, as well as food and nutrition. Investigations into advanced neuroimaging techniques for earlier and more precise characterization of molecular events at scales ranging from cellular to the full system level are equally important as it is necessary for the evaluation of the beneficial effects of different neuroprotective intervention approaches. AD, Alzheimer's disease; CNTF, ciliary neurotrophic factor; DHA, docosahexaenoic acid; GDF11, growth differentiation factor 11; GDNF, glial cell‐derived neurotrophic factor; IFN‐γ, interferon‐γ; NTFs, neurotrophic factors; ROS, reactive oxygen species; TNF‐α, tumor necrosis factor‐α.
1. INTRODUCTION
Alzheimer's disease (AD), the most common neurodegenerative disorder, is currently posing a threat to the health of approximately 35 million elderly individuals worldwide. 1 It was listed as the seventh‐leading cause of death in 2020. 2 The total estimated global socioeconomic burden of AD exceeds $1.3 trillion annually, and it is expected to double by 2030. 3 AD places a tremendous burden on not only individuals but also their families and the healthcare systems. This situation has led to an urgent requirement for efficient interventions against this devastating disease.
Accurate diagnosis and efficient treatment are two of the key aspects for AD intervention. Over the past two decades, several cohort studies have been launched to identify neuroimaging measures and biomarkers for AD diagnosis, 4 such as the Alzheimer's Disease Neuroimaging Initiative, the Australian Imaging, Biomarker & Lifestyle Flagship Study of Ageing, and the Dominantly Inherited Alzheimer Network. These studies facilitated the development of standardized methods for magnetic resonance imaging (MRI), positron emission tomography (PET), and cerebrospinal fluid (CSF) detection of AD biomarkers and promoted the establishment of the A (amyloid‐β [Aβ] deposition)—T (pathologic tau)—N (neurodegeneration) framework. 5 They shifted the definition of AD from a syndromic to a biological construct and provided a more reliable and accurate method for monitoring disease progression. Importantly, we are now capable of accurate and early AD diagnosis. Hence, it is now the time to focus our attention on early treatment strategies for patients diagnosed with AD.
2. CURRENT STATUS AND CHALLENGES OF AD TREATMENTS
The development of new AD therapies has always been challenging with the success rate of new drug development for AD being far below industry average. 6 Currently, approved pharmacological AD treatments are mainly divided into symptomatic drugs and disease‐modifying treatment (DMT) interventions. Symptomatic drugs, such as memantine, mainly work by cognitive enhancement or by controlling neuropsychiatric symptoms, without affecting the underlying biological causes of AD. These medications can only temporarily improve or maintain neuronal signaling and function, but they cannot reverse or halt AD progression. Therefore, there is an urgent need for DMT interventions that directly modify the AD‐specific pathobiological changes in the brain and exert neuroprotective effects, preventing diffuse neuronal cell loss. Recently, two DMT interventions, aducanumab, 7 and lecanemab, 8 both being monoclonal antibodies targeting brain Aβ plaques, have achieved some clinical success and have been approved by FDA for the treatment of mild cognitive impairment or mild dementia due to AD. Even though the long‐term safety and clinical efficacy of these two DMT interventions require further evaluation, their advent may indicate the dawn of a new era in AD therapy.
However, we still need to recognize that the current Aβ‐targeting approaches can only slow the progression of AD but cannot reverse or even halt the disease process. Therefore, additional interventions that can rescue cellular and circuit function in both early and advanced AD stages will be crucial for improving the patients' quality of life, and, therefore, should be the ultimate goal of therapeutic developments for AD. It is hence paramount for the global research community to focus attention on disease mechanisms and on how to stop or even reverse cognitive decline.
3. NEUROIMAGING FROM CELLULAR TO FULL SYSTEM LEVEL IN AD
Accurate diagnosis and efficient treatment are two key aspects of AD intervention with the aim of neuroprotection. Several cohort studies have allowed identification of neuroimaging measures and biomarkers for AD diagnosis, 4 demonstrating the capacity of these techniques for the evaluation of anatomic and functional changes in the brain. Moreover, advances in the field of nanotechnology have allowed extension of the capacities of these techniques, enabling them to become truly molecular imaging tools with which to visualize molecular events at the cellular to the full brain scale, allowing for a better understanding of AD pathophysiology. Thus, neuroimaging represents an invaluable asset in the development of novel therapies for the treatment of AD.
MRI is, possibly, the most versatile medical imaging technique available both at clinical and preclinical levels. Its noninvasiveness, image quality (in terms of image resolution and contrast generation), and multimodality allow detailed brain imaging at structural, vascular, metabolic, and functional levels. 9 In the field of AD, MRI is used to assess changes in brain structure, allowing the study of brain atrophy, 10 detecting events that may explain nondegenerative cognitive impairment, 11 and providing information about the integrity of white matter and neuronal circuits through all variants of diffusion‐weighted imaging (e.g., diffusion‐weighted imaging, diffusion tension imaging, diffusion kurtosis imaging, fiber tracking, neurite orientation dispersion, and density imaging, soma and neurite density imaging, etc. 12 ). Cerebrovascular alterations assessed through MRI‐based cerebral blood flow measurements (CBF‐MRI) can be used for the definition of early diagnosis of cognitive impairment, 13 along with magnetic resonance spectroscopy (1H‐MRS) or even more advanced spectroscopic approaches such as glutamate‐weighted chemical exchange saturation transfer imaging. This enables us to study metabolic alterations associated with AD progression. 14 One of the most valuable MRI modalities in AD is resting‐state functional MRI, which has been used to show how AD impairs multiple functional networks across the brain, including the default mode network, the salience network, or the dorsal attention network. 11
Nuclear imaging techniques such as PET and single photon emission computerized tomography are methods of choice to study important hallmarks of AD by using radioactive tracers that specifically bind to brain metabolites, protein aggregates, or neurotransmitters, or can visualize glucose metabolism, inflammatory events, and other processes involved in the AD pathophysiology. Nuclear medicine imaging techniques are extremely sensitive and can detect AD biomarkers at very early stages. PET is the gold standard technique for tau and Aβ‐specific imaging as well as for visualizing glucose metabolism in the brain. 11 Interestingly, PET has been used to show that tau may propagate along functional networks, instead of disseminating locally, but that this is not the case for amyloid. 15 Four different patterns for tau deposition have been described, potentially indicating AD subtypes, with different demographic and cognitive profiles, as well as outcomes. 16 However, these techniques lack the excellent spatial resolution and tissue contrast of MRI.
The recent introduction of powerful hybrid clinical imaging modalities, such as PET‐MRI allows us to capitalize on the advantages of each individual imaging modality while at the same overcoming their respective limitations. Using PET‐MRI, the A/T/N staging system has been defined, in which AD biomarkers are divided into three binary categories. In this system, A stands for β‐amyloid (measured in the brain by PET or alternatively using Aβ42 in CSF), T stands for tau (measured with specific PET tracers), and N stands for neurodegeneration or neuronal injury (determined by FDG‐PET and structural MRI).
Each of these three biomarkers is defined as positive or negative, and their combination results in a descriptive system for the categorization of the individual AD stage. These and other imaging‐based tools leaning on artificial intelligence, machine learning, deep neuronal networks, and other state‐of‐the‐art computing tools may ultimately advance to automated AD diagnosis and prognosis. 17 Thus, medical imaging applications in AD are a continuously evolving discipline, being of paramount importance.
4. INTERVENTIONS OF AD FROM THE PERSPECTIVE OF NEUROPROTECTION
Neurons in the AD brain undergo widespread degeneration, which ultimately results in cognitive impairment. Although Aβ deposition is the most important pathological hallmark of AD, the removal of Aβ from the brain through Aβ antibodies can only delay the progression of the disease, as it cannot reverse the neuronal loss (and related symptoms) that has occurred widely in the brain. Various cofactors such as genetics, inflammation, vascular issues, metabolism, and environment can influence neurodegeneration. We believe that only by targeting these pathological changes through different neuroprotective and potentially other therapeutic strategies can the cognitive function of AD patients be truly preserved or even restored (Table 1). Thus, holistic perspectives for neuroprotection strategies, focusing on the brain as well as the periphery, are discussed herein.
Table 1.
Targets of neuroprotective treatments for AD.
Targets | Evidence | References | |
---|---|---|---|
Central neuroprotective intervention | NTFs | Gene delivery of NGF into the basal forebrain shows promise in clinical trial in AD patients | [18] |
Antineuroinflammation | Pharmacological activation of microglial Piezo1 channels ameliorate brain Aβ burden and cognitive impairment | [19] | |
Replacement of reactive microglia in the aged brain reverses cognitive deficits in mice | [20] | ||
Astrocytic protection | Selective removal of astrocytic APOE4 protects against tau‐mediated neurodegeneration in PS19/APOE4 mice | [21] | |
Inhibition of the astrocytic protein S100B protects CA1 pyramidal neurons in AD mice model | [22] | ||
Remyelination | Genetically or pharmacologically enhancing myelin renewal reverses cognitive dysfunction in APP/PS1 mice | [23] | |
Facilitating oligodendrocytes cholesterol transport increases myelination and improves learning memory in APOE4/4‐TR mice | [24] | ||
Stem cell therapy and neural regeneration | Transplantation of NSCs increases adult hippocampal neurogenesis, rescues cognitive impairment in AD mice models | [25, 26] | |
Overexpression of NeuroD1 in reactive astrocytes of 5xFAD mice successfully converted the reactive astrocytes into functional neurons | [27] | ||
Glucose metabolism regulation | Administration of GLP‐1 agonist prevents memory impairments and synapse loss in APP/PS1 mice | [28] | |
Vascular system protection | Blocking RAGE at the BBB effectively reduces Aβ influx into the brain, and improves cognitive performance in APP/PS1 mice | [29] | |
Meningeal lymphatic protection | Augmentation of meningeal lymphatic drainage in aged mice improves cognitive function | [30] | |
Targeting prion‐like proteinopathy | Improving the O‐GlcNAcylation of tau prevents the development of tau pathology and functional deficits in PS19 mice | [31] | |
Peripheral neuroprotective intervention | Peripheral clearance of pathological substances | Enhancing the kidney's Aβ/tau clearing capacity either by peritoneal dialysis or pharmaceutical treatment mitigates cognitive impairment | [32, 33, 34] |
Stimulating Aβ uptake and metabolism by blood monocytes alleviates AD‐related pathology in APP/PS1 mice | [35] | ||
Peripheral neuroprotective factors | Exposing aged mice to blood from younger donors enhances hippocampal neurogenesis and boosts memory | [36, 37] | |
Neuroprotection by lifestyles changes | Physical exercise | Exercise plasma transfusion boosts memory and dampens brain inflammation | [38] |
Food and nutrition | Mediterranean diet and ketogenic diet show beneficial effects in AD patients | [39, 40] | |
Vitamin D, DHA, Ω‐3/Ω‐6, minerals, and phytochemical antioxidants supplementation exhibit neuroprotective effects in preclinical AD models and in clinical trials | [41, 42, 43, 44, 45] |
Abbreviations: Aβ, amyloid‐β; AD, Alzheimer's disease; BBB, blood–brain barrier; DHA, docosahexaenoic acid; GLP‐1, glucagon‐like peptide‐1; NGF, nerve growth factor; NSCs, neural stem cells; NTF, neurotrophic factor; RAGE, receptor for advanced glycation end products.
4.1. Central neuroprotective intervention
4.1.1. Neurotrophic factors (NTFs)
NTFs, such as neurotrophins and brain‐derived neurotrophic factor (BDNF), are endogenous soluble proteins supporting neurite outgrowth, neuronal cell differentiation, and survival. The dysregulation of NTFs and their receptors in AD is a key pathological process in the development of sporadic AD. 46 , 47 , 48 Loss in NTFs and their receptors also occurs with normal aging and is associated with normal cognitive decline, which seems to be accelerated in AD. The NTF ability to promote neuronal survival under pathological conditions has driven the idea that direct delivery of NTFs might be beneficial in AD. 49 Indeed, a gene therapy‐based approach for nerve growth factor delivery has shown promise in a clinical phase 1 trial in AD patients. 18 Instead of direct NTF delivery, the application of NTF receptor agonists could also exert neuroprotective effects. For instance, the BDNF mimetic compound 7,8‐dihydroxyflavone, a potent small molecular TrkB agonist, displays therapeutic efficacy against AD in mice. 50 , 51 Thus, the continued development of NTFs for therapy and the determination of whether this potent class of biologically active molecules will maintain the functional state of neurons in the AD brain are important lines of further investigation.
4.1.2. Antineuroinflammation
Large‐scale genome‐wide association studies (GWAS) indicate that about half of the AD risk genes are involved in immune processes. 52 This emphasizes the importance of immune dysfunction for AD development. Under physiological conditions, the immune function in the CNS is maintained in a dedicated balance to help the brain to detect and eliminate pathogens and cell debris without causing neuronal death. However, in the context of AD, neuroinflammation tends to be a chronic process that fails to resolve and is thought to accelerate disease progression. 53
Neuroinflammation is generally characterized by the production of proinflammatory cytokines by immune cells in the brain. The persistent or excessive release of proinflammatory molecules can lead to synaptic dysfunction, neuronal death, and finally cognitive impairment. 54 Therefore, being able to preserve cognitive function in AD will not be possible without controlling neuroinflammation. Currently, the number of clinical trials targeting neuroinflammation exceeds trials focusing on other mechanisms such as targeting Aβ, tau, and neurodegeneration. 55 Most of these trials applied broad‐spectrum anti‐inflammatory drugs to reduce inflammation, or antibodies to eliminate proinflammatory cytokines. However, none of these approaches targeted key regulators of neuroinflammation, that is, activated or dysregulated immune cells, and so they cannot fundamentally prevent neuroinflammation from occurring. This may be one of the reasons why current trials targeting neuroinflammation have not achieved clinical success.
Immune cells involved in neuroinflammation are mainly microglia and infiltrating monocytes/macrophages. There are also astrocytic contributions to neuroinflammation. In the process of AD, both microglia and astrocytes transit from a neurotrophic state to neurotoxic state, 56 but the underlying mechanism remains to be elucidated. 57 Recently, it was shown that a mechanotransduction ion channel, Piezo1, expressed in microglia orchestrates Aβ clearance by enhancing microglial survival, phagocytosis, and lysosomal activity. 58 Pharmacological activation of microglial Piezo1 ameliorated cerebral Aβ burden and cognitive impairment. 19 Replacement of reactive microglia also exhibited great potential in preserving cognitive function in aged mice. 20
Future investigations focusing on sensory mechanisms, downstream signaling pathways, and regulatory checkpoints of different immune cells in the brain will greatly advance our understanding of the contribution of neuroinflammation in AD pathogenesis and possibly help in identifying novel therapeutic targets.
4.1.3. Astrocytic protection
Astrocytes, which are the most abundant cell type in the CNS, play a crucial role in maintaining CNS homeostasis in terms of ion concentration balance, neurotransmitter buffering, synaptogenesis, stabilizing blood–brain barrier (BBB) function, and the secretion of neuroactive agents. In AD, however, astrocytes not only cease neurotrophic support but also secrete neurotoxic factors, which can accelerate disease progression. 56 , 59 Thus, enhancing the beneficial effects of astrocytes while dampening their negative properties shows great potential for disease modification in AD.
APOE4 is the strongest genetic risk factor for sporadic AD and is mainly expressed by astrocytes. 60 A recent study demonstrated that selective removal of astrocytic APOE4 strongly protects against tau‐mediated neurodegeneration. 21 Additionally, pentamidine, an inhibitor of the astrocytic protein S100B, has been shown to protect CA1 pyramidal neurons in a mouse model of AD by reducing expression of GFAP, S100B, and the receptor for advanced glycation end products (RAGE). 22 Other signaling cascades associated with astrocytes, such as signal transducer and activator of transcription 3, nuclear factor‐kappa B, and transforming growth factor beta, have also been implicated in AD pathology 56 , 61 and should be considered as putative targets for neuroprotection in future studies.
4.1.4. Remyelination
AD has been mainly considered a gray matter disorder; nevertheless, accumulating evidence emerging from imaging, postmortem, and genetic association studies suggests myelin impairment in AD. 62 , 63 The myelin sheath is a lipid‐rich multilamellar membrane that wraps around neuronal axons and thereby increases the conduction velocity of action potentials. In addition, it offers the necessary trophic support to the wrapped axons. Even though it is not clear whether alterations in myelination are directly involved in AD pathogenesis or just the secondary effect of neurodegeneration, enhancing myelin renewal was able to reverse cognitive dysfunction in AD mouse models. 23 Altered cholesterol localization coincides with reduced myelination in AD brains. Pharmacologically facilitating cholesterol transport has been shown to increase myelination and improve learning and memory in AD mice. 24 Therefore, future efforts to unveil the contribution of myelin dysfunction to AD and developing efficient myelination enhancement strategies require a greater effort in AD research.
4.1.5. Stem cell therapy and in vivo neural regeneration
Cognitive impairment in AD is due to neuronal degeneration and loss, leading to dysfunction and damage of neural circuits. Therefore, to improve cognitive function, it is necessary to replace lost neurons and repair damaged neural circuits. Hippocampal neurogenesis persists in the adult brain to maintain learning and memory, but is significantly reduced in AD. 64 Replenishing the exhausted pool of neural stem cells (NSCs) in the brain may help to rebuilt neural circuits and preserve cognitive function. Both in situ and noninvasive transplantation (like intranasal transplantation) of NSCs can increase adult hippocampal neurogenesis and mitigates cognitive impairment in AD mice. 25 , 26 However, repairing neuronal circuits in the mammalian brain can be challenging due to the absence of anatomical cues that guide brain development during embryogenesis and fetal development. One promising approach is the co‐transplantation of NSCs with bioengineered support cells that express target proteins. These support cells have the potential to produce growth factors that enhance the survival and migration of NSCs, eliminating the need for genetical engineering. 65 Several cell populations used in the treatment of neurodegenerative diseases have demonstrated neuroprotective benefits, 66 often referred to as “bystander effects”. These cells may also contribute to the prevention of neurodegeneration in AD. Another significant challenge in stem cell‐based therapy for AD is the difficulty of delivering stem cells in such a way that they can target all affected brain areas. Direct injection into the brain tissue provides limited coverage, 67 while intravenous administration is not effective with cells lost to filtering organs such as the lungs or liver, 68 and also the likelihood that only a few cells cross the BBB. A promising approach is to use the intra‐arterial route, which allows for more widespread and uniform distribution of cells to the targeted brain region. Imaging techniques that enable real‐time monitoring of cell distribution are of great interest, as they can increase precision and safety. 69 An alternative approach that is being investigated in preclinical studies is the use of stem cell‐derived extracellular vesicles (EVs). Several studies reported that the intranasal administration of mesenchymal stem cells‐derived EVs leads to improvements in mouse models of AD. 70 , 71
Instead of direct cell transplantation, a new technology has emerged for the regeneration of neurons through in vivo conversion of glial cells. 72 , 73 Specifically, the overexpression of the neural transcription factor NeuroD1 in reactive astrocytes of 5xFAD mice has successfully converted these astrocytes into functional neurons. 27 In addition to NeuroD1 and the AD mouse model, other transcription factors such as Ngn2, Ascl1, and Sox2 have been reported to convert astrocytes into neurons in various animal models, 74 , 75 , 76 including the first nonhuman primate stroke model. 77 Considering that human brain has 86 billion neurons, the loss of just 1% of neurons in the brain of an AD patient would amount to a loss of 860 million neurons. This extensive neuronal loss may explain why numerous drugs have failed to restore cognitive function in AD patients. Therefore, it may be crucial to combine neuroprotective strategies with in vivo neural regeneration to maximize the benefits for AD patients, particularly those in mid‐stage or even late‐stage AD.
4.1.6. Cerebral glucose metabolism regulation
The brain is the most energy‐demanding organ in the human body. Even though it constitutes only 2% of the total body weight, it accounts for 20% of an individual's energy expenditure. 78 Insufficient energy supply to the brain swiftly results in irreversible impairment of brain function. 79 The major energy source for the human brain is glucose; unlike in peripheral organs, the brain has a very limited availability of other sources of energy (such as fatty acids). Glucose hypometabolism is one of the earliest pathologic events in AD. 80 Reduced glucose metabolism in the brain may be caused by the decreased expression of glucose transporters, 81 decreased activities of enzymes involved in glucose metabolism, 82 and disrupted insulin/insulin‐like growth factor (IGF) signaling pathway. 83 Among them, the insulin/IGF signaling pathway is currently most intensively studied. In addition to participating in cerebral bioenergetics regulation, the insulin/IGF signaling pathway also contributes to Aβ metabolism, tau phosphorylation, and neuroinflammation. 84 Thus, AD is proposed to be type III diabetes, and this concept has emerged as a very promising area of AD research.
An intriguing therapeutic target is the glucagon‐like peptide‐1 receptor (GLP‐1R). Glucagon is an endogenous insulinotropic hormone that participates in the homeostatic regulation of insulin and glucose. The activation of the GLP‐1Rs affects neuronal excitability, synaptic plasticity, and memory processes, and GLP‐1 analogs have been successfully tested in both preclinical models of neurodegeneration and clinical trials. 28 , 85 Future efforts should be made to develop more effective approaches to restore cerebral glucose metabolism, thereby offering a therapeutic benefit to AD patients.
4.1.7. Cerebral blood vessel protection
Cerebral blood vessels and the BBB function as the gatekeepers for the shuttling of ions, molecules, and cells between the blood and the brain, protecting the brain from peripheral toxins and pathogens. 86 They also mediate the clearance of brain‐derived neurotoxins such as Aβ and pTau, as well as metabolic waste, into circulation. A recent single‐nuclear transcriptome profiling study in human brain vascular and perivascular cells revealed that 30 of the top 45 AD risk genes identified by GWAS analysis are expressed in the cerebral vasculature, 87 which further supports the link between the vascular system and AD. Neuroimaging studies indicate that loss of BBB integrity is an early sign of AD and may contribute to cognitive dysfunction in AD. 88 , 89 Postmortem studies show that the vast majority of patients diagnosed with AD invariably bear some degree of cerebral vascular pathology, such as cerebral small vessel disease (cSVD). 90 Conditions typically associated with cSVD such as hypertension may increase RAGE expression at the BBB, fostering Aβ influx to the brain, and accelerating cerebral Aβ deposition. 91 Blocking RAGE can effectively reduce Aβ influx and improve cognitive performance in APP/PS1 mice. 29 Altogether, these pieces of evidence indicate that there is potential in targeting the vascular system for AD treatment. However, even though existing studies in both animal models and patients have confirmed that the BBB plays a key role in AD etiology, it is yet to be explored as a therapeutic target. 92 Thus, future studies focusing on restoring the impaired BBB, targeting the BBB clearance machinery, or eliminating the consequences of BBB breakdown 93 might promote drug research and discovery for AD.
4.1.8. Meningeal lymphatic protection
The brain has traditionally been regarded as an immune‐privileged organ 94 due to the limited entry of immune cells into the healthy brain parenchyma. However, several groups have recently discovered and characterized lymphatic vessels within the meninges. 95 , 96 It is now widely recognized that meningeal lymphatic vessels play an essential role in maintaining brain homeostasis by draining neurotoxic substances (such as Aβ) from the brain into the cervical lymph nodes, 97 and lymphatic vessel dysfunction may be one of the underlying factors for worsened Aβ pathology and cognitive deficits in AD. 98 Moreover, impaired meningeal lymphatic drainage could exacerbate the microglial inflammatory response in AD. 99 , 100 In addition, abnormal lymphatic function can affect the effectiveness of immunotherapy targeting Aβ. The ablation of meningeal lymphatic vessels in 5xFAD mice worsened the outcome after treatment with mouse chimeric analogs of aducanumab. 99 The above evidence suggests that lymphatic vessels play an important role in both the pathogenesis and prognosis of AD and represent an important target for AD treatment. Indeed, augmentation of meningeal lymphatic drainage in 5xFAD mice can facilitate the clearance of macromolecules from the brain, resulting in improved cognitive function. 30 Future studies should investigate the mechanisms that drive lymphatic vessel dysfunction, and develop feasible interventions to improve brain lymphatic drainage and ameliorate the progression of AD.
4.1.9. Targeting prion‐like proteinopathy
Tau pathology has been strongly correlated with ante mortem cognitive decline in AD patients. 101 , 102 , 103 α‐Synuclein (α‐syn) pathology is present in 30%–50% of AD patients, and is associated with a more rapid cognitive decline and more severe neuropsychiatric dysfunction. 104 , 105 Heiko Braak and his colleagues discovered tau and α‐syn pathology exhibiting clear progressive and hierarchical spreading patterns in postmortem brains. 106 , 107 , 108 , 109 Both clinical and experimental observations support that inoculation with pathologic tau and α‐syn can induce substantial prion‐like protein aggregation 110 , 111 , 112 resulting in neurodegeneration and cognitive impairment. 113 , 114 This indicates that prion‐like tau and α‐syn spreading are both major triggers and drivers. Multiple pathways/mechanisms are involved in the cell‐to‐cell transmission of pathogenic prion‐like seeds, including receptors, 115 , 116 cell death, 117 inflammation, 118 oxidative stress, 119 aggregation clearance, 120 and strain mediation. 121 , 122 All of those are potential therapeutic targets.
Uptake of prion‐like seeds via receptors is required to initiate subsequent prion propagation and neurotoxicity. Genetic depletion and inhibition of these receptors effectively block cell uptake, prion propagation, neurodegeneration, and behavioral deficits. 115 , 116 , 123 , 124 After uptake, prion‐like seeds can induce reactive oxygen species (ROS), activate poly (adenosine 5′‐diphosphate‐ribose) polymerase‐1 (PARP‐1), and cause DNA damage. Inhibition of PARP and depletion of PARP‐1 can significantly alleviate prion propagation and associated neurodegeneration. 117 Not only neuronal cells but also glial cells play critical roles in these pathological processes. Inhibiting microglial activation induced by seeds can substantially reduce the activation of astrocytes and associated neurotoxicity. 118 Prion‐like seeds can induce oxidative stress, which in turn induces further protein aggregation. This vicious circle drives disease progression. Nanozymes can strongly scavenge ROS induced by prion‐like seeds, inhibit a feed‐forward loop, and block prion‐like seed propagation in vivo. 119 Tau and α‐syn pathology are observed intracellularly; however, it is challenging to develop agents that can target intracellular pathogenic seeds. Nanobodies capable of specifically recognizing and clearing the fibrillar form of prion‐like seeds were developed, and adeno‐associated virus‐based gene delivery methods provide an attractive approach to inhibit the propagation of prion‐like seeds in preclinical models. 31 , 120 Diverse misfolded aggregates with distinct molecular conformations (strains) are a prion feature that causes a different disease phenotype. The strategies targeting strain mediation, such as inhibiting the strain inducer (i.e., poly adenosine 5′‐diphosphate‐ribose 117 ) or adding strain modulator (i.e., glucocerebrosidase 120 ), may provide an effective impact against prion‐like propagation and neurotoxicity.
4.2. Peripheral neuroprotective intervention
As outlined above, the development of AD is not caused by a single factor but involves multisystem and multilevel changes. 125 Many peripheral factors are involved in AD development, such as immune senescence, 126 metabolic 127 and cardiovascular 128 disorders, liver and kidney dysfunction, 32 gut microbiota disturbance, 129 or respiratory and sleep disorders. 130 Therefore, controlling system comorbidity and improving whole‐body health will positively impact brain structure and function.
4.2.1. Peripheral clearance of neurotoxic substances
Aβ and pathological tau have long been considered to be cleared by central metabolic pathways. But in fact, both brain‐derived Aβ and tau can be cleared after being transported to peripheral organs and cells, 33 , 131 such as the liver, 131 kidney, 32 spleen, 132 and blood monocytes. 133 This suggests that improving peripheral Aβ/tau clearance capacity could be a desirable therapeutic strategy for AD. For instance, enhancing the kidney's Aβ/tau clearing capacity through peritoneal dialysis 33 , 34 or pharmaceutical treatment 32 has been shown to mitigate cognitive impairment in AD mice. Stimulating monocytic Aβ uptake and metabolism through polysaccharide krestin, a modulator of innate immune signaling, could also alleviate AD‐related pathology in AD mouse models. 35 Plasma exchange has shown potential in slowing the rate of cognitive decline in AD patients in clinical trials as well. 134 However, these concepts are still being explored in fundamental research, and future translational and clinical studies are necessary to evaluate the effectiveness and safety of this strategy in AD treatment.
4.2.2. Systemic neuroprotective factors
Peripheral organs secrete NTFs to boost neurogenesis and reduce neuronal apoptosis, as indicated in heterochronic parabiosis models. 135 The circulatory system integrates signals from all organs and provides a route for crosstalk between peripheral organs/tissues and the brain. For example, exposing aged mice to blood from younger donors enhances hippocampal neurogenesis, induces immediate to early gene activation, increases dendritic spine density, and promotes vascular remodeling. 36 , 37
However, due to the potential risks, it is unrealistic to directly apply young plasma transfusion for AD treatment. Hence, the identification of substances in young plasma such as peripheral NTFs that can exert neuroprotective effects is a subject worth further studying. Current research is mainly focused on plasma proteomics and several proteins that promote neurogenesis have been identified, including growth differentiation factor 11, metalloproteinase 2, colony‐stimulating factor 2, and osteocalcin. 136 , 137
Apart from the identification of neurotrophic proteins, the identification of potential neuroprotective metabolites in youthful plasma is equally important. Moreover, the investigation of exosomes in the plasma is also very promising. Exosomes are membrane‐bound, secreted organelles, and are enriched in proteins, metabolites, and nucleic acids, which can reflect their cells of origin. Due to its single‐membrane structure, the substances in the exosome are more stable than in plasma. Thus, proteomics, metabolomics, and miRNAomics studies on young plasma exosomes are promising approaches for the identification of new neuroprotective substances.
4.3. Neuroprotection by lifestyle changes
4.3.1. Physical exercise
It is now widely accepted that physical exercise can combat neurodegeneration and provide neuroprotection in elderly individuals. Both animal and clinical experiments have demonstrated that physical exercise enhances the proliferation and differentiation of NSCs, promoting NTFs secretion, and finally improving cognitive performance. 138
However, little is known about the molecular mechanisms of how exercise affects brain health. It has been proposed that “exercise factors,” secreted from muscle and other tissues into the blood, mediate this beneficial effect. A recent study found that transfusions of plasma obtained after exercise boosts memory and dampens brain inflammation. 38 Further studies showed that exercise can promote the secretion of myokines from muscles. 139 These myokines were then released into blood and cross the BBB to promote the expression of NTFs in the brain. 140 Animal experiments indicate that peripheral overexpression of myokines is sufficient to promote cognitive function. 141 Furthermore, liver metabolism also changes drastically during exercise. A recent study found that β‐hydroxybutyrate produced by the liver during exercise promotes BDNF expression. 142 There is also evidence that exercise can improve cognitive function by promoting cerebral angiogenesis. 143 However, many growth factors need to reach a threshold level of expression to exert beneficial effects. This indicates that conventional behavioral/exercise interventions may not be of benefit as they are too short to trigger a significant increase in growth factors. 144
In general, research in this area is still in its infancy. Future studies should focus on identifying new “exercise factors” that may improve physical exercise regimens (or exert effects similar to it) for elderly patients with AD and disabilities who cannot perform strenuous exercise.
4.3.2. Food and nutrition
Nutrition is important for the health and well‐being of people living with AD, as weight loss and malnutrition are important complications in AD patients. 145 Thus, maintaining and improving nutritional status in AD patients may help to delay AD development by promoting brain health. Some dietary patterns have been shown to be beneficial in AD, such as the Mediterranean diet 39 and the ketogenic diet. 40 In addition, certain active substances in the diet (for instance, vitamin D, 41 docosahexaenoic acid, 42 omega‐3 and omega‐6, 43 minerals, 44 and phytochemical antioxidants 45 ) were identified as neuroprotective factors. As nutrition‐related conditions can occur in all stages of AD, further basic and clinical studies are recommended to elucidate the exact mechanism of malnutrition and weight loss in AD patients and whether nutritional intervention can improve cognitive symptoms.
5. SUMMARY
Over the last years, several clinical trials were launched trying to overcome AD, and three antibody‐based DMT interventions targeting Aβ have achieved limited clinical success by slowing down the pace of AD in early stages. However, merely slowing down the decline of cognitive function is not sufficient to achieve the clinical goal of AD treatment, which is to preserve the patient's cognitive function. As widespread neurodegeneration in the brain forms the biological basis of the patient's cognitive impairment, developing neuroprotective strategies should become the next important direction of AD treatment after Aβ clearance (Figure 1). However, this field has not garnered sufficient attention thus far, and the progress made has been limited. Furthermore, AD encompasses pathological alterations in numerous facets. Consequently, the objectives of future research lie in the precise classification of AD patients and the application of distinct combinations of intervention strategies tailored to individual patients.
Figure 1.
Neuroprotective interventions for AD. Main neuroprotective intervention targets for AD, including NTFs, antineuroinflammation, astrocytic protection, remyelination, stem‐cell therapy and in vivo neural regeneration, glucose metabolism regulation, vascular system protection, lymphatic protection, prion‐like proteinopathy regulation, peripheral neurotoxic substances clearance, systemic NTFs, and physical exercise, as well as food and nutrition. Investigations into advanced neuroimaging techniques for earlier and more precise characterization of molecular events at scales ranging from cellular to full system level are equally important as it is necessary for the evaluation of the beneficial effects of different neuroprotective intervention approaches. AD, Alzheimer's disease; CNTF, ciliary neurotrophic factor; DHA, docosahexaenoic acid; GDF11, growth differentiation factor 11; GDNF, glial cell‐derived neurotrophic factor; IFN‐γ, interferon‐γ; NTFs, neurotrophic factors; ROS, reactive oxygen species; TNF‐α, tumor necrosis factor‐α.
Therefore, we convened a group of experts to jointly issue this initiative to emphasize the importance of neuroprotection research. ADNRI mainly focuses on the prevention of AD, which aims to prevent the loss of neurons and the destruction of neural networks in the early stage of the disease through various neuroprotective strategies. We call on our colleagues around the globe to cooperate to reveal the mechanisms of neurological deficits in AD and identify effective neuroprotective targets, to carry out translational research, and to jointly achieve the goal of preserving the cognitive function of AD patients.
AUTHOR CONTRIBUTIONS
Jie Liu: Resources (equal); writing—original draft (equal). Heleen van Beusekom: Conceptualization (equal); writing—review and editing (lead). Xian‐Le Bu: Conceptualization (equal); writing—review and editing (equal). Gong Chen: Conceptualization (equal); writing—review and editing (equal). Paulo Henrique Rosado de Castro: Conceptualization (equal); writing—review and editing (equal). Xiaochun Chen: Conceptualization (equal); writing—review and editing (equal). Xiaowei Chen: Conceptualization (equal); writing—review and editing (equal). Andrew N. Clarkson: Conceptualization (equal); writing—review and editing (equal). Tracy D. Farr: Conceptualization (equal); writing—review and editing (equal). Yuhong Fu: Conceptualization (equal); writing—review and editing (equal). Jianping Jia: Conceptualization (equal); writing—review and editing (equal). Jukka Jolkkonen: Conceptualization (equal); writing—review and editing (equal). Woojin Scott Kim: Conceptualization (equal); writing—review and editing (equal). Paula Korhonen: Conceptualization (equal); writing—review and editing (equal). Shen Li: Conceptualization (equal); writing—review and editing (equal). Yajie Liang: Conceptualization (equal); writing—review and editing (equal). Guang‐Hui Liu: Conceptualization (equal); writing—review and editing (equal). Guiyou Liu: Conceptualization (equal); writing—review and editing (equal). Yu‐Hui Liu: Conceptualization (equal); writing—review and editing (equal). Tarja Malm: Conceptualization (equal); writing—review and editing (equal). Xiaobo Mao: Conceptualization (equal); writing—review and editing (equal). Joaquim Miguel Oliveira: Conceptualization (equal); writing—review and editing (equal). Mike M. Modo: Conceptualization (equal); writing—review and editing (equal). Pedro Ramos‐Cabrer: Conceptualization (equal); writing—review and editing (equal). Karsten Ruscher: Conceptualization (equal); writing—review and editing (equal). Weihong Song: Conceptualization (equal); writing—review and editing (equal). Jun Wang: Conceptualization (equal); writing—review and editing (equal). Xuanyue Wang: Conceptualization (equal); writing—review and editing (equal). Yun Wang: Conceptualization (equal); writing—review and editing (equal). Haitao Wu: Conceptualization (equal); writing—review and editing (equal). Lize Xiong: Conceptualization (equal); writing—review and editing (equal). Yi Yang: Conceptualization (equal); writing—review and editing (equal). Keqiang Ye: Conceptualization (equal); writing—review and editing (equal). Jin‐Tai Yu: Conceptualization (equal); writing—review and editing (equal). Xin‐Fu Zhou: Conceptualization (equal); writing—review and editing (equal). Marietta Zille: Conceptualization (equal); writing—review and editing (equal). Colin L. Masters: Conceptualization (equal); writing—review and editing (equal). Piotr Walczak: Conceptualization (lead); funding acquisition (lead); writing—review and editing (lead). Johannes Boltze: Conceptualization (lead); funding acquisition (lead); writing—review and editing (lead). Xunming Ji: Conceptualization (lead); funding acquisition (lead); writing—review and editing (lead). Yan‐Jiang Wang: Conceptualization (lead); funding acquisition (lead); writing—original draft (equal); writing—review and editing (lead).
CONFLICT OF INTEREST STATEMENT
Xunming Ji, Johannes Boltze, and Piotr Walczak are the Editors‐in‐Chief, and Heleen van Beusekom, Andrew N. Clarkson, Paulo Henrique Rosado de Castro, Tracy D. Farr, Jukka Jolkkonen, Shen Li, Yajie Liang, Guiyou Liu, Xiaobo Mao, Joaquim Miguel Oliveira, Mike M. Modo, Pedro Ramos‐Cabrer, Karsten Ruscher, Yun Wang, Haitao Wu, Lize Xiong, Yi Yang, Marietta Zille, and Yan‐Jiang Wang are the editorial members, of Neuroprotection. They are therefore excluded from the peer‐review process and all editorial decisions related to the publication of this manuscript. The remaining authors declare no conflict of interest.
ETHICS STATEMENT
The authors have nothing to report.
ACKNOWLEDGMENTS
Piotr Walczak is a co‐founder and co‐owner of IntraART, LLC and Ti‐Com, LLC. Yan‐Jiang Wang is supported by the National Natural Science Foundation of China (92249305, 82120108010, 81930028, 31921003). Johannes Boltze is supported by funding from The Academy of Medical Sciences (Newton Advanced Fellowship NAF/R11/1010). Piotr Walczak is supported by funding from the National Institutes of Health (No. R01DA056739).
Liu J, van Beusekom H, Bu X‐L, et al. Preserving cognitive function in patients with Alzheimer's disease: the Alzheimer's disease neuroprotection research initiative (ADNRI). Neuroprotection. 2023;1:84‐98. 10.1002/nep3.23
Contributor Information
Piotr Walczak, Email: pwalczak@som.umaryland.edu.
Johannes Boltze, Email: johannes.boltze@warwick.ac.uk.
Xunming Ji, Email: jixm@ccmu.edu.cn.
Yan‐Jiang Wang, Email: yanjiang_wang@tmmu.edu.cn.
DATA AVAILABILITY STATEMENT
The authors have nothing to report.
REFERENCES
- 1. WHO . Dementia . 2022. Accessed May 20, 2023. https://www.who.int/news-room/fact-sheets/detail/dementia
- 2. WHO . The Top 10 Causes of Death . 2020. Accessed May 20, 2023. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death
- 3. World Alzheimer Report . 2022. Life after Diagnosis: Navigating Treatment, Care and Support. 2022. Accessed May 20, 2023.
- 4. Mueller SG, Weiner MW, Thal LJ, et al. Ways toward an early diagnosis in Alzheimer's disease: the Alzheimer's disease neuroimaging initiative (ADNI). Alzheimer's Dement. 2005;1(1):55‐66. 10.1016/j.jalz.2005.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Jack CR Jr., Bennett DA, Blennow K, et al. NIA‐AA research framework: toward a biological definition of Alzheimer's disease. Alzheimer's Dement. 2018;14(4):535‐562. 10.1016/j.jalz.2018.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Calcoen D, Elias L, Yu X. What does it take to produce a breakthrough drug? Nat Rev Drug Discovery. 2015;14(3):161‐162. 10.1038/nrd4570 [DOI] [PubMed] [Google Scholar]
- 7. Sevigny J, Chiao P, Bussière T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature. 2016;537(7618):50‐56. 10.1038/nature19323 [DOI] [PubMed] [Google Scholar]
- 8. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in early Alzheimer's disease. N Engl J Med. 2023;388(1):9‐21. 10.1056/NEJMoa2212948 [DOI] [PubMed] [Google Scholar]
- 9. Huang P, Zhang M. Magnetic resonance imaging studies of neurodegenerative disease: from methods to translational research. Neurosci Bull. 2023;39(1):99‐112. 10.1007/s12264-022-00905-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Elliott ML, Hanford LC, Hamadeh A, et al. Brain morphometry in older adults with and without dementia using extremely rapid structural scans. Neuroimage. 2023;276:120173. 10.1016/j.neuroimage.2023.120173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Risacher SL, Apostolova LG. Neuroimaging in dementia. Continuum (Minneap Minn). 2023;29(1):219‐254. 10.1212/con.0000000000001248 [DOI] [PubMed] [Google Scholar]
- 12. Andica C, Kamagata K, Hatano T, et al. MR biomarkers of degenerative brain disorders derived from diffusion imaging. J Magn Reson Imaging. 2020;52(6):1620‐1636. 10.1002/jmri.27019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kapasouri EM, Ioannidis DC, Cameron D, Vassiliou VS, Hornberger M. The utility of arterial spin labeling MRI in medial temporal lobe as a vascular biomarker in Alzheimer's disease spectrum: a systematic review and meta‐analysis. Diagnostics. 2022;12(12):2967. 10.3390/diagnostics12122967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. McKiernan E, Su L, O'Brien J. MRS in neurodegenerative dementias, prodromal syndromes and at‐risk states: a systematic review of the literature. NBM. 2023;36(7):e4896. 10.1002/nbm.4896 [DOI] [PubMed] [Google Scholar]
- 15. Vogel JW, Iturria‐Medina Y, Strandberg OT, et al. Spread of pathological tau proteins through communicating neurons in human Alzheimer's disease. Nat Commun. 2020;11(1):2612. 10.1038/s41467-020-15701-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Vogel JW, Young AL, Oxtoby NP, et al. Four distinct trajectories of tau deposition identified in Alzheimer's disease. Nat Med. 2021;27(5):871‐881. 10.1038/s41591-021-01309-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Eslami M, Tabarestani S, Adjouadi M. A unique color‐coded visualization system with multimodal information fusion and deep learning in a longitudinal study of Alzheimer's disease. Artif Intell Med. 2023;140:102543. 10.1016/j.artmed.2023.102543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Tuszynski MH, Yang JH, Barba D, et al. Nerve growth factor gene therapy: activation of neuronal responses in Alzheimer disease. JAMA Neurol. 2015;72(10):1139‐1147. 10.1001/jamaneurol.2015.1807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Hu J, Chen Q, Zhu H, et al. Microglial Piezo1 senses Aβ fibril stiffness to restrict Alzheimer's disease. Neuron. 2023;111(1):15‐29.e8. 10.1016/j.neuron.2022.10.021 [DOI] [PubMed] [Google Scholar]
- 20. Elmore MRP, Hohsfield LA, Kramár EA, et al. Replacement of microglia in the aged brain reverses cognitive, synaptic, and neuronal deficits in mice. Aging Cell. 2018;17(6):e12832. 10.1111/acel.12832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wang C, Xiong M, Gratuze M, et al. Selective removal of astrocytic APOE4 strongly protects against tau‐mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron. 2021;109(10):1657‐1674.e7. 10.1016/j.neuron.2021.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Cirillo C, Capoccia E, Iuvone T, et al. S100B inhibitor pentamidine attenuates reactive gliosis and reduces neuronal loss in a mouse model of Alzheimer's disease. BioMed Res Int. 2015;2015:1‐11. 10.1155/2015/508342 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Chen JF, Liu K, Hu B, et al. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer's disease. Neuron. 2021;109(14):2292‐2307.e5. 10.1016/j.neuron.2021.05.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Blanchard JW, Akay LA, Davila‐Velderrain J, et al. APOE4 impairs myelination via cholesterol dysregulation in oligodendrocytes. Nature. 2022;611(7937):769‐779. 10.1038/s41586-022-05439-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lu MH, Ji WL, Chen H, et al. Intranasal transplantation of human neural stem cells ameliorates Alzheimer's disease‐like pathology in a mouse model. Front Aging Neurosci. 2021;13:650103. 10.3389/fnagi.2021.650103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhang HA, Yuan CX, Liu KF, et al. Neural stem cell transplantation alleviates functional cognitive deficits in a mouse model of tauopathy. Neural Regen Res. 2022;17(1):152‐162. 10.4103/1673-5374.314324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Guo Z, Zhang L, Wu Z, Chen Y, Wang F, Chen G. In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer's disease model. Cell Stem Cell. 2014;14(2):188‐202. 10.1016/j.stem.2013.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Bassil F, Fernagut PO, Bezard E, Meissner WG. Insulin, IGF‐1 and GLP‐1 signaling in neurodegenerative disorders: targets for disease modification? Prog Neurobiol. 2014;118:1‐18. 10.1016/j.pneurobio.2014.02.005 [DOI] [PubMed] [Google Scholar]
- 29. Deane R, Singh I, Sagare AP, et al. A multimodal RAGE‐specific inhibitor reduces amyloid β‐mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest. 2012;122(4):1377‐1392. 10.1172/jci58642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Choi C, Park J, Kim H, Chang KT, Park J, Min KT. DSCR1 upregulation enhances dural meningeal lymphatic drainage to attenuate amyloid pathology of Alzheimer's disease. J Pathol. 2021;255(3):296‐310. 10.1002/path.5767 [DOI] [PubMed] [Google Scholar]
- 31. Permanne B, Sand A, Ousson S, et al. O‐GlcNAcase inhibitor ASN90 is a multimodal drug candidate for tau and α‐synuclein proteinopathies. ACS Chem Neurosci. 2022;13(8):1296‐1314. 10.1021/acschemneuro.2c00057 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Tian DY, Cheng Y, Zhuang ZQ, et al. Physiological clearance of amyloid‐beta by the kidney and its therapeutic potential for Alzheimer's disease. Mol Psychiatry. 2021;26(10):6074‐6082. 10.1038/s41380-021-01073-6 [DOI] [PubMed] [Google Scholar]
- 33. Wang J, Jin WS, Bu XL, et al. Physiological clearance of tau in the periphery and its therapeutic potential for tauopathies. Acta Neuropathol. 2018;136(4):525‐536. 10.1007/s00401-018-1891-2 [DOI] [PubMed] [Google Scholar]
- 34. Jin WS, Shen LL, Bu XL, et al. Peritoneal dialysis reduces amyloid‐beta plasma levels in humans and attenuates Alzheimer‐associated phenotypes in an APP/PS1 mouse model. Acta Neuropathol. 2017;134(2):207‐220. 10.1007/s00401-017-1721-y [DOI] [PubMed] [Google Scholar]
- 35. Chen SH, He CY, Shen YY, et al. Polysaccharide krestin prevents Alzheimer's disease‐type pathology and cognitive deficits by enhancing monocyte amyloid‐β processing. Neurosci Bull. 2021;38(3):290‐302. 10.1007/s12264-021-00779-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Villeda SA, Plambeck KE, Middeldorp J, et al. Young blood reverses age‐related impairments in cognitive function and synaptic plasticity in mice. Nat Med. 2014;20(6):659‐663. 10.1038/nm.3569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Katsimpardi L, Litterman NK, Schein PA, et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science. 2014;344(6184):630‐634. 10.1126/science.1251141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. De Miguel Z, Khoury N, Betley MJ, et al. Exercise plasma boosts memory and dampens brain inflammation via clusterin. Nature. 2021;600(7889):494‐499. 10.1038/s41586-021-04183-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Ballarini T, Melo van Lent D, Brunner J, et al. Mediterranean diet, Alzheimer disease biomarkers and brain atrophy in old age. Neurology. 2021;96(24):e2920‐e2932. 10.1212/wnl.0000000000012067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Broom GM, Shaw IC, Rucklidge JJ. The ketogenic diet as a potential treatment and prevention strategy for Alzheimer's disease. Nutrition. 2019;60:118‐121. 10.1016/j.nut.2018.10.003 [DOI] [PubMed] [Google Scholar]
- 41. Jia J, Hu J, Huo X, Miao R, Zhang Y, Ma F. Effects of vitamin D supplementation on cognitive function and blood Aβ‐related biomarkers in older adults with Alzheimer's disease: a randomised, double‐blind, placebo‐controlled trial. J Neurol Neurosurg Psychiatry. 2019;90:jnnp‐2018‐320199. 10.1136/jnnp-2018-320199 [DOI] [PubMed] [Google Scholar]
- 42. Zhang YP, Lou Y, Hu J, Miao R, Ma F. DHA supplementation improves cognitive function via enhancing Aβ‐mediated autophagy in Chinese elderly with mild cognitive impairment: a randomised placebo‐controlled trial. J Neurol Neurosurg Psychiatry. 2018;89(4):382‐388. 10.1136/jnnp-2017-316176 [DOI] [PubMed] [Google Scholar]
- 43. Cutuli D, De Bartolo P, Caporali P, et al. n‐3 polyunsaturated fatty acids supplementation enhances hippocampal functionality in aged mice. Front Aging Neurosci. 2014;6:220. 10.3389/fnagi.2014.00220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Wang ZX, Tan L, Wang HF, et al. Serum iron, zinc, and copper levels in patients with Alzheimer's disease: a replication study and meta‐analyses. J Alzheimer's Dis. 2015;47(3):565‐581. 10.3233/jad-143108 [DOI] [PubMed] [Google Scholar]
- 45. Jiao SS, Yao XQ, Liu YH, et al. Edaravone alleviates Alzheimer's disease‐type pathologies and cognitive deficits. Proc Natl Acad Sci USA. 2015;112(16):5225‐5230. 10.1073/pnas.1422998112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Yao XQ, Jiao SS, Saadipour K, et al. p75NTR ectodomain is a physiological neuroprotective molecule against amyloid‐beta toxicity in the brain of Alzheimer's disease. Mol Psychiatry. 2015;20(11):1301‐1310. 10.1038/mp.2015.49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Weinstein G, Beiser AS, Choi SH, et al. Serum brain‐derived neurotrophic factor and the risk for dementia: the framingham heart study. JAMA Neurol. 2014;71(1):55‐61. 10.1001/jamaneurol.2013.4781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Ginsberg SD, Malek‐Ahmadi MH, Alldred MJ, et al. Brain‐derived neurotrophic factor (BDNF) and TrkB hippocampal gene expression are putative predictors of neuritic plaque and neurofibrillary tangle pathology. Neurobiol Dis. 2019;132:104540. 10.1016/j.nbd.2019.104540 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Xu YL, Zhu L, Chen ZJ, et al. Release of endogenous brain‐derived neurotrophic factor into the lateral entorhinal cortex from the paraventricular thalamus ameliorates social memory deficits in a mouse model of Alzheimer's disease. Neurosci Bull. 2022;38(11):1425‐1430. 10.1007/s12264-022-00900-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Chen C, Wang Z, Zhang Z, et al. The prodrug of 7,8‐dihydroxyflavone development and therapeutic efficacy for treating Alzheimer's disease. Proc Natl Acad Sci USA. 2018;115(3):578‐583. 10.1073/pnas.1718683115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Jang SW, Liu X, Yepes M, et al. A selective TrkB agonist with potent neurotrophic activities by 7,8‐dihydroxyflavone. Proc Natl Acad Sci USA. 2010;107(6):2687‐2692. 10.1073/pnas.0913572107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Bellenguez C, Küçükali F, Jansen IE, et al. New insights into the genetic etiology of Alzheimer's disease and related dementias. Nat Genet. 2022;54(4):412‐436. 10.1038/s41588-022-01024-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here. Nat Rev Neurol. 2021;17(3):157‐172. 10.1038/s41582-020-00435-y [DOI] [PubMed] [Google Scholar]
- 54. Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflammation: the role and consequences. Neurosci Res. 2014;79:1‐12. 10.1016/j.neures.2013.10.004 [DOI] [PubMed] [Google Scholar]
- 55. Cummings J, Lee G, Nahed P, et al. Alzheimer's disease drug development pipeline: 2022. Alzheimers Dement (N Y). 2022;8(1):e12295. 10.1002/trc2.12295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Monterey MD, Wei H, Wu X, Wu JQ. The many faces of astrocytes in Alzheimer's disease. Front Neurol. 2021;12:619626. 10.3389/fneur.2021.619626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Zhang L, Wang Y, Liu T, Mao Y, Peng B. Novel microglia‐based therapeutic approaches to neurodegenerative disorders. Neurosci Bull. 2023;39(3):491‐502. 10.1007/s12264-022-01013-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Jäntti H, Sitnikova V, Ishchenko Y, et al. Microglial amyloid beta clearance is driven by PIEZO1 channels. J Neuroinflammation. 2022;19(1):147. 10.1186/s12974-022-02486-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Brandebura AN, Paumier A, Onur TS, Allen NJ. Astrocyte contribution to dysfunction, risk and progression in neurodegenerative disorders. Nat Rev Neurosci. 2022;24(1):23‐39. 10.1038/s41583-022-00641-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15(9):501‐518. 10.1038/s41582-019-0228-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Rodríguez‐Giraldo M, González‐Reyes RE, Ramírez‐Guerrero S, Bonilla‐Trilleras CE, Guardo‐Maya S, Nava‐Mesa MO. Astrocytes as a therapeutic target in Alzheimer's disease‐comprehensive review and recent developments. Int J Mol Sci. 2022;23(21):13630. 10.3390/ijms232113630 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nasrabady SE, Rizvi B, Goldman JE, Brickman AM. White matter changes in Alzheimer's disease: a focus on myelin and oligodendrocytes. Acta Neuropathol Commun. 2018;6(1):22. 10.1186/s40478-018-0515-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Qu Y, Wang P, Yao H, et al. Reproducible abnormalities and diagnostic generalizability of white matter in Alzheimer's disease. Neurosci Bull. 2023. 10.1007/s12264-023-01041-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Navarro Negredo P, Yeo RW, Brunet A. Aging and rejuvenation of neural stem cells and their niches. Cell Stem Cell. 2020;27(2):202‐223. 10.1016/j.stem.2020.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Liang Y, Ågren L, Lyczek A, Walczak P, Bulte JWM. Neural progenitor cell survival in mouse brain can be improved by co‐transplantation of helper cells expressing bFGF under doxycycline control. Exp Neurol. 2013;247:73‐79. 10.1016/j.expneurol.2013.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Janowski M, Wagner DC, Boltze J. Stem cell‐based tissue replacement after stroke: factual necessity or notorious fiction? Stroke. 2015;46(8):2354‐2363. 10.1161/strokeaha.114.007803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Li S, Oh BC, Chu C, et al. Induction of immunological tolerance to myelinogenic glial‐restricted progenitor allografts. Brain. 2019;142(11):3456‐3472. 10.1093/brain/awz275 [DOI] [PubMed] [Google Scholar]
- 68. Lappalainen RS, Narkilahti S, Huhtala T, et al. The SPECT imaging shows the accumulation of neural progenitor cells into internal organs after systemic administration in middle cerebral artery occlusion rats. Neurosci Lett. 2008;440(3):246‐250. 10.1016/j.neulet.2008.05.090 [DOI] [PubMed] [Google Scholar]
- 69. Walczak P, Wojtkiewicz J, Nowakowski A, et al. Real‐time MRI for precise and predictable intra‐arterial stem cell delivery to the central nervous system. J Cereb Blood Flow Metab. 2017;37(7):2346‐2358. 10.1177/0271678x16665853 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Cone AS, Yuan X, Sun L, et al. Mesenchymal stem cell‐derived extracellular vesicles ameliorate Alzheimer's disease‐like phenotypes in a preclinical mouse model. Theranostics. 2021;11(17):8129‐8142. 10.7150/thno.62069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Losurdo M, Pedrazzoli M, D'Agostino C, et al. Intranasal delivery of mesenchymal stem cell‐derived extracellular vesicles exerts immunomodulatory and neuroprotective effects in a 3xTg model of Alzheimer's disease. Stem Cells Transl Med. 2020;9(9):1068‐1084. 10.1002/sctm.19-0327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Li H, Chen G. In vivo reprogramming for CNS repair: regenerating neurons from endogenous glial cells. Neuron. 2016;91(4):728‐738. 10.1016/j.neuron.2016.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Bocchi R, Masserdotti G, Götz M. Direct neuronal reprogramming: fast forward from new concepts toward therapeutic approaches. Neuron. 2022;110(3):366‐393. 10.1016/j.neuron.2021.11.023 [DOI] [PubMed] [Google Scholar]
- 74. Mattugini N, Bocchi R, Scheuss V, et al. Inducing different neuronal subtypes from astrocytes in the injured mouse cerebral cortex. Neuron. 2019;103(6):1086‐1095.e5. 10.1016/j.neuron.2019.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Su Z, Niu W, Liu ML, Zou Y, Zhang CL. In vivo conversion of astrocytes to neurons in the injured adult spinal cord. Nat Commun. 2014;5(1):3338. 10.1038/ncomms4338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Rao Z, Wang R, Li S, et al. Molecular mechanisms underlying Ascl1‐mediated astrocyte‐to‐neuron conversion. Stem Cell Reports. 2021;16(3):534‐547. 10.1016/j.stemcr.2021.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Ge LJ, Yang FH, Li W, et al. In vivo neuroregeneration to treat ischemic stroke through neurod1 AAV‐based gene therapy in adult non‐human primates. Front Cell Dev Biol. 2020;8:590008. 10.3389/fcell.2020.590008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Hyder F, Rothman DL, Bennett MR. Cortical energy demands of signaling and nonsignaling components in brain are conserved across mammalian species and activity levels. Proc Natl Acad Sci USA. 2013;110(9):3549‐3554. 10.1073/pnas.1214912110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Zilberter Y, Zilberter M. The vicious circle of hypometabolism in neurodegenerative diseases: ways and mechanisms of metabolic correction. J Neurosci Res. 2017;95(11):2217‐2235. 10.1002/jnr.24064 [DOI] [PubMed] [Google Scholar]
- 80. Ewers M, Brendel M, Rizk‐Jackson A, et al. Reduced FDG‐PET brain metabolism and executive function predict clinical progression in elderly healthy subjects. NeuroImage: Clinical. 2014;4:45‐52. 10.1016/j.nicl.2013.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Winkler EA, Nishida Y, Sagare AP, et al. GLUT1 reductions exacerbate Alzheimer's disease vasculo‐neuronal dysfunction and degeneration. Nat Neurosci. 2015;18(4):521‐530. 10.1038/nn.3966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Sorbi S, Bird ED, Blass JP. Decreased pyruvate dehydrogenase complex activity in Huntington and Alzheimer brain. Ann Neurol. 1983;13(1):72‐78. 10.1002/ana.410130116 [DOI] [PubMed] [Google Scholar]
- 83. Stanley M, Macauley SL, Holtzman DM. Changes in insulin and insulin signaling in Alzheimer's disease: cause or consequence? J Exp Med. 2016;213(8):1375‐1385. 10.1084/jem.20160493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kellar D, Craft S. Brain insulin resistance in Alzheimer's disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020;19(9):758‐766. 10.1016/S1474-4422(20)30231-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. McClean PL, Parthsarathy V, Faivre E, Hölscher C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer's disease. J Neurosci. 2011;31(17):6587‐6594. 10.1523/jneurosci.0529-11.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Barisano G, Montagne A, Kisler K, Schneider JA, Wardlaw JM, Zlokovic BV. Blood‐brain barrier link to human cognitive impairment and Alzheimer's disease. Nat Cardiovasc Res. 2022;1(2):108‐115. 10.1038/s44161-021-00014-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Yang AC, Vest RT, Kern F, et al. A human brain vascular Atlas reveals diverse mediators of Alzheimer's risk. Nature. 2022;603(7903):885‐892. 10.1038/s41586-021-04369-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Montagne A, Nation DA, Sagare AP, et al. APOE4 leads to blood‐brain barrier dysfunction predicting cognitive decline. Nature. 2020;581(7806):71‐76. 10.1038/s41586-020-2247-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sweeney MD, Montagne A, Sagare AP, et al. Vascular dysfunction‐the disregarded partner of Alzheimer's disease. Alzheimer's Dement. 2019;15(1):158‐167. 10.1016/j.jalz.2018.07.222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Kalaria RN, Sepulveda‐Falla D. Cerebral small vessel disease in sporadic and familial Alzheimer disease. Am J Pathol. 2021;191(11):1888‐1905. 10.1016/j.ajpath.2021.07.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Carnevale D, Mascio G, D'Andrea I, et al. Hypertension induces brain β‐amyloid accumulation, cognitive impairment, and memory deterioration through activation of receptor for advanced glycation end products in brain vasculature. Hypertension. 2012;60(1):188‐197. 10.1161/hypertensionaha.112.195511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Govindpani K, Vinnakota C, Waldvogel H, Faull R, Kwakowsky A. Vascular dysfunction in Alzheimer's disease: a biomarker of disease progression and a potential therapeutic target. Neural Regen Res. 2020;15(6):1030‐1032. 10.4103/1673-5374.270306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Paul J, Strickland S, Melchor JP. Fibrin deposition accelerates neurovascular damage and neuroinflammation in mouse models of Alzheimer's disease. J Exp Med. 2007;204(8):1999‐2008. 10.1084/jem.20070304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Hašek M, Chutna J, Sládeček M, Lodin Z. Immunological tolerance and tumour allografts in the brain. Nature. 1977;268(5615):68‐69. 10.1038/268068a0 [DOI] [PubMed] [Google Scholar]
- 95. Louveau A, Smirnov I, Keyes TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337‐341. 10.1038/nature14432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Absinta M, Ha SK, Nair G, et al. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. eLife. 2017;6:e29738. 10.7554/eLife.29738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Li X, Qi L, Yang D, et al. Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat Neurosci. 2022;25(5):577‐587. 10.1038/s41593-022-01063-z [DOI] [PubMed] [Google Scholar]
- 98. Da Mesquita S, Louveau A, Vaccari A, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer's disease. Nature. 2018;560(7717):185‐191. 10.1038/s41586-018-0368-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Da Mesquita S, Papadopoulos Z, Dykstra T, et al. Meningeal lymphatics affect microglia responses and anti‐Aβ immunotherapy. Nature. 2021;593(7858):255‐260. 10.1038/s41586-021-03489-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Louveau A, Herz J, Alme MN, et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci. 2018;21(10):1380‐1391. 10.1038/s41593-018-0227-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Ossenkoppele R, Lyoo CH, Jester‐Broms J, et al. Assessment of demographic, genetic, and imaging variables associated with brain resilience and cognitive resilience to pathological tau in patients with Alzheimer disease. JAMA Neurol. 2020;77(5):632‐642. 10.1001/jamaneurol.2019.5154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. La Joie R, Visani AV, Baker SL, et al. Prospective longitudinal atrophy in Alzheimer's disease correlates with the intensity and topography of baseline tau‐PET. Sci Transl Med. 2020;12(524):eaau5732. 10.1126/scitranslmed.aau5732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Tanner JA, Iaccarino L, Edwards L, et al. Amyloid, tau and metabolic PET correlates of cognition in early and late‐onset Alzheimer's disease. Brain. 2022;145(12):4489‐4505. 10.1093/brain/awac229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Olichney JM, Galasko D, Salmon DP, et al. Cognitive decline is faster in Lewy body variant than in Alzheimer's disease. Neurology. 1998;51(2):351‐357. 10.1212/wnl.51.2.351 [DOI] [PubMed] [Google Scholar]
- 105. Chung EJ, Babulal GM, Monsell SE, Cairns NJ, Roe CM, Morris JC. Clinical features of Alzheimer disease with and without Lewy bodies. JAMA Neurol. 2015;72(7):789‐796. 10.1001/jamaneurol.2015.0606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Braak H, Braak E. Staging of Alzheimer's disease‐related neurofibrillary changes. Neurobiol Aging. 1995;16(3):271‐278. 10.1016/0197-4580(95)00021-6 [DOI] [PubMed] [Google Scholar]
- 107. Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. Staging of Alzheimer disease‐associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006;112(4):389‐404. 10.1007/s00401-006-0127-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Braak H, Tredici KD, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. 2003;24(2):197‐211. 10.1016/s0197-4580(02)00065-9 [DOI] [PubMed] [Google Scholar]
- 109. Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host‐to‐graft disease propagation. Nat Med. 2008;14(5):501‐503. 10.1038/nm1746 [DOI] [PubMed] [Google Scholar]
- 110. Guo JL, Narasimhan S, Changolkar L, et al. Unique pathological tau conformers from Alzheimer's brains transmit tau pathology in nontransgenic mice. J Exp Med. 2016;213(12):2635‐2654. 10.1084/jem.20160833 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Bassil F, Meymand ES, Brown HJ, et al. α‐Synuclein modulates tau spreading in mouse brains. J Exp Med. 2021;218(1):e20192193. 10.1084/jem.20192193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Narasimhan S, Guo JL, Changolkar L, et al. Pathological tau strains from human brains recapitulate the diversity of tauopathies in nontransgenic mouse brain. J Neurosci. 2017;37(47):11406‐11423. 10.1523/jneurosci.1230-17.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. He Z, Guo JL, McBride JD, et al. Amyloid‐β plaques enhance Alzheimer's brain tau‐seeded pathologies by facilitating neuritic plaque tau aggregation. Nat Med. 2018;24(1):29‐38. 10.1038/nm.4443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Bassil F, Brown HJ, Pattabhiraman S, et al. Amyloid‐beta (Aβ) plaques promote seeding and spreading of alpha‐synuclein and tau in a mouse model of Lewy body disorders with Aβ pathology. Neuron. 2020;105(2):260‐275.e6. 10.1016/j.neuron.2019.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Mao X, Ou MT, Karuppagounder SS, et al. Pathological α‐synuclein transmission initiated by binding lymphocyte‐activation gene 3. Science. 2016;353(6307):aah3374. 10.1126/science.aah3374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Mao X, Gu H, Kim D. Aplp1 and the Aplp1‐Lag3 complex facilitates transmission of pathologic α‐synuclein. bioRxiv. 2021;2021.2005.2001.442157. 10.1101/2021.05.01.442157 [DOI] [Google Scholar]
- 117. Kam TI, Mao X, Park H, et al. Poly(ADP‐ribose) drives pathologic α‐synuclein neurodegeneration in Parkinson's disease. Science. 2018;362(6414):eaat8407. 10.1126/science.aat8407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Yun SP, Kam TI, Panicker N, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease. Nat Med. 2018;24(7):931‐938. 10.1038/s41591-018-0051-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Liu Y‐Q, Mao Y, Xu E, et al. Nanozyme scavenging ROS for prevention of pathologic α‐synuclein transmission in Parkinson's disease. Nano Today. 2021;36:101027. 10.1016/j.nantod.2020.101027 [DOI] [Google Scholar]
- 120. Seo BA, Kim D, Hwang H, et al. TRIP12 ubiquitination of glucocerebrosidase contributes to neurodegeneration in Parkinson's disease. Neuron. 2021;109(23):3758‐3774.e11. 10.1016/j.neuron.2021.09.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Jia L, Liu Y, Wang W, et al. Molecular mediation of prion‐like α‐synuclein fibrillation from toxic PFFs to nontoxic species. ACS Appl Bio Mater. 2020;3(9):6096‐6102. 10.1021/acsabm.0c00684 [DOI] [PubMed] [Google Scholar]
- 122. Li A, Rastegar C, Mao X. α‐Synuclein conformational plasticity: physiologic states, pathologic strains, and biotechnological applications. Biomolecules. 2022;12(7):994. 10.3390/biom12070994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Zhang S, Liu YQ, Jia C, et al. Mechanistic basis for receptor‐mediated pathological α‐synuclein fibril cell‐to‐cell transmission in Parkinson's disease. Proc Natl Acad Sci USA. 2021;118(26):e2011196118. 10.1073/pnas.2011196118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Gu H, Yang X, Mao X, et al. Lymphocyte activation gene 3 (Lag3) contributes to α‐synucleinopathy in α‐synuclein transgenic mice. Front Cell Neurosci. 2021;15:656426. 10.3389/fncel.2021.656426 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease—insights from amyloid‐β metabolism beyond the brain. Nat Rev Neurol. 2017;13(10):612‐623. 10.1038/nrneurol.2017.111 [DOI] [PubMed] [Google Scholar]
- 126. Cao W, Zheng H. Peripheral immune system in aging and Alzheimer's disease. Mol Neurodegener. 2018;13(1):51. 10.1186/s13024-018-0284-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity. 2022;55(1):31‐55. 10.1016/j.immuni.2021.12.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Tublin JM, Adelstein JM, Del Monte F, Combs CK, Wold LE. Getting to the heart of Alzheimer disease. Circ Res. 2019;124(1):142‐149. 10.1161/circresaha.118.313563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Wang X, Sun G, Feng T, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids‐shaped neuroinflammation to inhibit Alzheimer's disease progression. Cell Res. 2019;29(10):787‐803. 10.1038/s41422-019-0216-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Irwin MR, Vitiello MV. Implications of sleep disturbance and inflammation for Alzheimer's disease dementia. Lancet Neurol. 2019;18(3):296‐306. 10.1016/s1474-4422(18)30450-2 [DOI] [PubMed] [Google Scholar]
- 131. Xiang Y, Bu XL, Liu YH, et al. Physiological amyloid‐beta clearance in the periphery and its therapeutic potential for Alzheimer's disease. Acta Neuropathol. 2015;130(4):487‐499. 10.1007/s00401-015-1477-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Yu ZY, Chen DW, Tan CR, et al. Physiological clearance of Aβ by spleen and splenectomy aggravates Alzheimer‐type pathogenesis. Aging cell. 2022;21(1):e13533. 10.1111/acel.13533 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Chen SH, Tian DY, Shen YY, et al. Amyloid‐beta uptake by blood monocytes is reduced with ageing and Alzheimer's disease. Transl Psychiatry. 2020;10(1):423. 10.1038/s41398-020-01113-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Boada M, López OL, Olazarán J, et al. A randomized, controlled clinical trial of plasma exchange with albumin replacement for Alzheimer's disease: primary results of the AMBAR study. Alzheimer's Dement. 2020;16(10):1412‐1425. 10.1002/alz.12137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Villeda SA, Luo J, Mosher KI, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477(7362):90‐94. 10.1038/nature10357 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Castellano JM, Mosher KI, Abbey RJ, et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature. 2017;544(7651):488‐492. 10.1038/nature22067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Khrimian L, Obri A, Ramos‐Brossier M, et al. Gpr158 mediates osteocalcin's regulation of cognition. J Exp Med. 2017;214(10):2859‐2873. 10.1084/jem.20171320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Erickson KI, Voss MW, Prakash RS, et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci USA. 2011;108(7):3017‐3022. 10.1073/pnas.1015950108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Dalise S, Cavalli L, Ghuman H, et al. Biological effects of dosing aerobic exercise and neuromuscular electrical stimulation in rats. Sci Rep. 2017;7(1):10830. 10.1038/s41598-017-11260-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Moon HY, Becke A, Berron D, et al. Running‐induced systemic cathepsin b secretion is associated with memory function. Cell Metab. 2016;24(2):332‐340. 10.1016/j.cmet.2016.05.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Lourenco MV, Frozza RL, de Freitas GB, et al. Exercise‐linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer's models. Nat Med. 2019;25(1):165‐175. 10.1038/s41591-018-0275-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Sleiman SF, Henry J, Al‐Haddad R, et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β‐hydroxybutyrate. eLife. 2016;5:e15092. 10.7554/eLife.15092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Ding YH, Li J, Zhou Y, Rafols J, Clark J, Ding Y. Cerebral angiogenesis and expression of angiogenic factors in aging rats after exercise. Curr Neurovasc Res. 2006;3(1):15‐23. 10.2174/156720206775541787 [DOI] [PubMed] [Google Scholar]
- 144. Berretta A, Tzeng YC, Clarkson AN. Post‐stroke recovery: the role of activity‐dependent release of brain‐derived neurotrophic factor. Expert Rev Neurother. 2014;14(11):1335‐1344. 10.1586/14737175.2014.969242 [DOI] [PubMed] [Google Scholar]
- 145. Díaz G, Lengele L, Sourdet S, Soriano G, de Souto Barreto P. Nutrients and amyloid β status in the brain: A narrative review. Ageing Res Rev. 2022;81:101728. 10.1016/j.arr.2022.101728 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The authors have nothing to report.