• Open Access
    Review

    Looking at the periphery—new hypothesis to look for new targets for Alzheimer’s disease therapy

    Jesús Avila 1,2*
    Mar Pérez 3
    Marina Avila-Villanueva 4
    Ismael Santa-María 5
    Félix Hernández 1

    Explor Neuroprot Ther. 2023;3:151–163 DOI: https://doi.org/10.37349/ent.2023.00044

    Received: December 02, 2022 Accepted: March 17, 2023 Published: June 30, 2023

    Academic Editor: Rafael Franco, Universidad de Barcelona, Spain

    This article belongs to the special issue The Urgent Need for New Hypotheses to Develop Effective Therapeutic Tools Against Alzheimer's Disease

    Abstract

    Currently, the predominant targets for the treatment of Alzheimer’s disease (AD) are the main components of the two pathological structures: senile plaques (composed of amyloid beta peptide aggregates) or neurofibrillary tangles (constructed of tau protein polymers). However, the existence of adequate disease modifiers based on such targets is discussed. In this special issue, it has been suggested to search for new possible targets for AD therapy. This contribution tries to analyze non-neuronal tissues (periphery) to identify potential factors (target) involved in the development of AD.

    Keywords

    Alzheimer’s disease, non-neuronal tissues, biomarkers

    Introduction

    A strategy to prevent a disease is to determine the primary cause of that disease. In the case of Alzheimer’s disease (AD), there are two main types: familial AD (FAD) and sporadic AD (SAD). It is mainly assumed that the primary cause for FAD is the presence of mutations in some genes like presenilin 1 (PSEN1), presenilin 2 (PSEN2) , precursor of beta amyloid peptide (APP) [1] and, probably, sortilin related receptor 1 (SORL1) [2, 3] that could favor the presence of beta amyloid (Aβ) peptide aggregates, being that presence the first step for AD development. In this way, FAD could be similar to other neurodegenerative disorders, like Huntington’s disease (HD), that results from a genetic mutation [4]. However, even assuming that it was the primary cause, there is not suitable therapies for FAD or HD.

    For FAD, the great majority of the cases are the consequences of PSEN1 gene mutations and it has been described that those PSEN1 mutations may produce alterations appearing in the brain previously to Aβ over-production, in a mouse FAD model [5]. Thus, the presence of Aβ could not be the first step in FAD due to mutated PSEN1. Nevertheless, the prevalent view is that in FAD, the increase in Aβ is a very early factor in the development of the disease and that it could be, also, for SAD (amyloid hypothesis) [6].

    However, in SAD, there are other additional hypotheses suggesting primary causes for the disease [7]. Examples of those hypotheses are: cholinergic [8], calcium [9], oxidative stress [10], neuron inflammation [11], microbial (viral) infections [12], glucose hypometabolism [13] or the presence of an excess of tau aggregates [14], among others.

    On the other hand, the main risk factor for SAD is aging [15] and healthy longevity could prevent the appearance of SAD.

    Aging can be observed at different levels: the whole organism, organs (or tissues), cellular, and molecular levels. At cellular level, after the cells become old could be an additional step, senescence, with the senescence-associated secretion phenotype (SASP) [16] in which senescent cells could secrete toxic substances to the surrounding cells, with or without the same origin. Thus, the presence of a tissue with senescent cells could facilitate the toxicity and aging of neighboring tissues. Recently, it has been described an example, showing that skin cell aging drives age related bone loss via secreted cystatin A [17], or that knee osteoarthritis (OA), in old people, may accelerate Aβ deposition and neurodegeneration in the central nervous system (CNS) [18], although the presence of the brain blood barrier (BBB) may difficult the crossing of secreted compounds from the periphery to CNS. In any way, it has been proposed that AD may be a multiorgan disorder [19].

    In addition to being a neurodegenerative disorder, AD pathology may include immune system dysfunction [20], inflammation, dysbiosis, insulin resistance, heart problems, kidney, and hepatic dysfunction, among others [19].

    However, the main pathology is related to CNS disorders and the disorders in other tissues and organs may be used, or not, as putative biomarkers of the disease, although further work should be done to know if those peripheral disorders are cause or consequence of the CNS.

    CNS disorders at cellular level

    The most well-known neuronal hallmarks in AD are senile plaques [21] and neurofibrillary tangles [22], coming from two proteins, APP and tau protein, mainly expressed in neurons [2325]. In addition, glia cells, present in CNS, may play an important role in the development of AD. In this way, microglia can have harmful actions in AD [26]. Activated microglia or disease associated microglia (DAM) with the new nomenclature [27] may be one of the key factors involved in the neuroinflammation found in AD [28]. Also, the activation of microglia could be the consequence of a change in the cross-talking neuron-resting microglia [29] and could be the consequence of microglia change of status specific features found in AD mouse models. Parabiosis experiments in mouse models indicate that microglia account for the increased myeloid cell number observed in brains with plaque pathology [30]. Additionally, astrocytes may also play a role in neuroinflammation [31].

    Is there any relation between peripheral nervous system and AD?

    AD is a CNS disorder but it could correlate with some changes in the peripheral nervous system (PNS) that could be used as biomarkers of the disease. An example is the lower degree of pain observed in AD patients [32].

    About peripheral biomarkers, in human fluids, in addition to looking at the levels of Aβ or tau protein in cerebral spinal fluid (CSF) [33, 34], other peripheral fluids, like plasma, saliva, or urine, have been used as possible markers of compounds, like proinflammatory cytokines, or others, that could be altered in AD patients [35]. The kinetics for the appearance of these markers could be important since earlier markers could facilitate the earlier prevention of the disease [35].

    Peripheral disorders related to AD found in patients or in animal models

    Briefly, it will be described some peripheral features that are present in AD patients or in models for the disease, although, probably, in many cases the features could be more the consequence than the cause, that facilitate the development of the disorders. On the other hand, the use of palliative pharmacological treatments, for the disease, could result in damage to organs like kidney or liver.

    Here, it is commented on the possible relation of AD development with seven different organs: heart, blood, gut, muscle, bone, kidney, and liver (Figure 1). Although other organs like lungs have been suggested to get a link with dementia [36], little is known about molecular factors involved in that interaction and they are not mentioned here.

    Peripheral disorders related to AD. The figure showed different organs that could express or secrete compounds associated with features found in AD (like Aβ aggregates, proinflammatory cytokines, insulin dysfunction, or small fat-derived metabolites) or compounds that could act as protective factors (like irisin)

    Heart

    Links between cardiovascular abnormalities and neurological disorders in AD patients have been reported [37] suggesting that “mind the heart in Alzheimer disease” [38, 39]. Also, a long time ago, the term “cardiogenic dementia” was indicated [40], being, actually, a possible therapeutical treatment, systolic blood pressure intervention trial memory and cognition in decreased hypertension (SPRINTMIND), based on the fact that a healthy brain starts with a healthy heart and that control of intensive blood pressure could reduce dementia [41].

    Blood

    Aging, but mainly a development of AD, results, for example, in a decline of adult hippocampal neurogenesis and cognitive function [42]. To avoid that aging cognitive decline, heterochronic parabiosis, mixing the blood of young and old mice, could help the recovery of cognitive function in old mice [43]. The levels of some compounds, like the chemokine eotaxin, are increased in old mice or aging humans, and those compounds could impair memory or cognitive functions [43]. Possible therapies based on blood exchange have been proposed as a possible treatment of neurodegenerative disorders [43]. Also, related to AD, it has been described that clearance of brain Aβ burden may take place by blood exchange, suggesting that defects in peripheral circulation may play a role in AD pathogenesis [44]. In this way, it has been indicated that blood-derived amyloid peptides could induce AD-like pathology [45] and behavioral deficits [46].

    Gut

    About the role of brain-gut microbiota axis dysregulation, many works have been published. Indeed, a whole long review article should deserve this subject, but, in this review, only a brief comment should be included to indicate the gut changes related to brain pathogenesis [47]. Changes in gut microbiota (dysbiosis) may induce increased permeability of the intestinal barrier leading to systemic inflammation. Curiously, Aβ has been described as an antimicrobial peptide [48] that could participate in that immune response [49]. It should be noted that intestinal inflammation triggers C/EBPβ/δ-secretase-dependent spread from gut to brain of Aβ in AD, suggesting that the gut is likely a critical source of Aβ in the brain [50]. In addition, there is a gut-derived amyloid that may contribute to Alzheimer’s pathogenesis [51]. Also, intra-gastrointestinal Aβ oligomers perturb enteric function and may facilitate the development of AD pathology found in another mouse model [52].

    On the other hand, a high proportion of patients with inflammatory bowel disease show mental disorders, perhaps due to changes in brain choroid plexus modulated in response to intestinal inflammation [53]. Additionally, immunotherapy targeting tau can modulate gut microbiota in a mouse model for AD [54].

    The presence of specific bacteria strains in gut microbiota could facilitate or decrease serotonin secretion and could promote neurotransmission dysfunction [55], which could result in depression, a risk factor for AD [56, 57].

    The level of other metabolites like butyrate or propionic acid [58] should be also modulated by gut microbiota. By dietary source of tryptophan, some bacteria types could be recruited to the gut, increasing serotonin levels [59].

    On the other hand, microbiota-dependent progression of neurodegeneration in a mouse model of tauopathies has been reported [60].

    Finally, linking with muscle functions (see below), gut microbes shape athletic motivation [61], that could favor physical exercise, a protective factor for dementia.

    Muscle

    As previously indicated, depression is a risk factor of dementia and decreased levels of brain-derived neurotrophic factor (BNDF) are related to depression, but there is a peptide, irisin, induced by physical (muscular) exercise that increases the level of BNDF and cognition in mouse models for AD. Recently, low levels of irisin have been found in CSF from old humans with depression [62].

    Irisin is a cleaved peptide from transmembrane fibronectin type II domain-containing protein 5 (FNDC5) that has been linked to adult neurogenesis, being a possible mediator of muscle-brain crosstalk, that could explain the therapy based on physical exercise for patients with dementia [63]. In any case, irisin may be suggested as a possible biomarker for dementia [62].

    Liver

    It has been suggested that Aβ peptides produced in the liver can be involved in the formation of senile plaques [64]. Based on that, it has been proposed that peripheral (liver) Aβ clearance could be a good therapy for AD [6567].

    In addition, an association of altered liver enzymes with the diagnosis of AD has been proposed, implying changes in cognition, neuroimaging measures, or in the level of cerebrospinal fluid markers [68]. In addition, brain changes related to hepatic apolipoprotein E4 (ApoE4) expression have been reported in mice with humanized livers [69]. These changes are probably due to the role of peripheral ApoE4 in cognition impairment, by compromising cerebrovascular function [70]. Indeed, liver-restricted expression of human amyloid results in an AD-like neurodegenerative phenotype [71].

    Bone

    OA and AD are two main genetic diseases that could have common features like the presence of chronic inflammation [72]. Also, the prevalence of dementia in OA patients is higher than in non-OA subjects and treatment with etoricoxib and diclofenac may reduce the risk of dementia in patients with OA [73].

    A possible link between OA and AD is that OA accelerates amyloid beta deposition and neurodegeneration, as it has been shown in a mouse model of AD [18].

    Kidney

    Physiological clearance of the Aβ peptide by the kidney has been reported [74]. On the other hand, tau, the main component of neurofibrillary tangles in AD, is usually considered as a specific neuronal protein [12] but its possible presence in peripheral tissues was proposed a long time ago [75]. However, recent studies have indicated that tau could be present in kidney, playing a role in the morphology of podocyte architecture [76]. Also, in some tauopathies, like frontotemporal dementia, some patients report urinary incontinence [77] and, more recently, the association of impaired kidney functions with dementia and brain pathologies has been indicated [78, 79]. Curiously, AD patients with chronic kidney disease (CKD) treated with cholinesterase inhibitors showed a lower risk of CKD [80]. Due to this link between brain and kidney pathologies and formic acid, a toxic compound associated with several cognitive disorders, may be present in the urine of AD patients. It was found an increase in formic acid level measured in the urine of AD patients and it could be used as a biomarker for the dementia and used as a diagnostic tool in the future [81].

    Pancreas

    Insulin is a peptide hormone expressed in the beta cells present in pancreas. Insulin has a role in the regulation of the metabolism of carbohydrates, lipid, and proteins through the absorption of glucose from blood to some organs. The action of insulin is through its receptors present in different cell types [82].

    Insulin receptors are expressed in cells from both PNS and CNS [83], playing some functions related, for example, to food intake or memory consolidation [84].

    Defects in insulin function may result in disorders such as diabetes mellitus. There are two types of this disorder, type I and II. In type I, no insulin is made and in type II, the insulin pathway initiated by the binding of the hormone to its cellular receptors is not working and, as a consequence of that, the expressed insulin does not have the desired functional effect in the body.

    Diabetes mellitus type II accounts for more than 90% of all diabetes mellitus cases and a correlation between type II diabetes and dementia has been reported, with diabetes being an important risk factor for the development of AD [84, 85]. Thus, some authors have described AD as diabetes type III [86].

    New targets, therapies or biomarkers for AD

    Actually, the main targets for AD therapies are the components of senile plaques, Aβ peptide aggregates; or the component of neurofibrillary tangles, tau protein, since the amyloid [6] and tau protein [14] hypothesis are now the most popular hypothesis for AD development. Mainly, antibodies against Aβ aggregates have been tested, as a possible therapy for amyloid pathology. Although some of those antibodies like aducanumab or lecanemab have been raised some good expectations, no suitable results were obtained and, in addition, some side effects, like cerebral hemorrhages, were found in some treated patients [87]. More recently, it has been also indicated an accelerated brain volume loss is caused by these anti Aβ drugs [88]. Indeed, almost all of clinical trials for AD have failed to show positive results as indicated in the commentary “The long road to a cure for AD is paved with failures” recently published in Nature Medicine [89], discussing the last data in clinical trials for multiple targets like Aβ, tau, inflammation components, and other ones, see also [90]. Also, Aβ and tau have been mainly used as early biomarkers for AD, looking at fluids like CSF, or blood, see for example [91]. Also, from glia (microglia) cells, triggering receptor expressed on myeloid cells 2 (TREM2) level could be associated with the transition from mild cognitive impairment to dementia, during the development of the AD continuum process [92].

    On the other hand, it has been discussed why successful results have been obtained in animal models but not in humans [93]. Also, some compounds that could result in a neurodegeneration or cognitive improvement have been proposed as possible palliative therapies [94].

    The use of other compounds related to some features found in AD has been reported. For example, the small compound “simufilam”, a stabilizer of filamin A [95], has been proposed as a possible therapy for AD, although that use is under discussion [96].

    Now, new targets or new early biomarkers for AD are being looked for. About new possible biomarkers for AD, from the periphery, some examples related to blood are included. A possible link between platelet biomarkers and cognitive functions has been proposed [97] and between plasma β2 microglobulin and N-methyl-D-aspartate (NMDA) receptors, since β2 microglobulin could act as an antagonist for NMDA receptors, resulting in cognitive impairment [98]. Also, related to blood, associations of plasma cluster of differentiation 22 (CD22) level with cognitive decline in AD have been reported [99].

    Biomarkers in other fluids like saliva and urine, for AD diagnosis, have been also proposed. Since chronic stress is a risk factor for the disease, the level of cortisol (related to stress) could be used as a prodromic marker for the AD continuum process [100]. Additionally, as indicated in kidney section, the presence of formic acid in urine could be used as a possible marker for dementia [81].

    Conclusions

    This brief review is mainly focused on the contribution of peripheral tissues in AD development, although, in many cases, the observed peripheral pathology could be more the consequence than a possible cause for AD.

    It is well-known that changes in life style, or improving the quality of nutrition, could result in a decrease in the incidence of AD development [101, 102]. Treatment of risk factors like hypertension or diabetes or playing physical exercise could retard the appearance of dementia, decreasing the fast progression of AD at world level [103]. Additionally, since aging is the main risk factor for neurodegenerative disorders, a possible therapy to reprogram (rejuvenate) old into young neurons has been proposed [104]. Also, the presence of senescent cells, like senescent astrocytes, with SASP, may contribute to an increase in Aβ aggregation or tau phosphorylation [105] and, in that way, the use of senolytic drugs has been proposed in clinical trials for AD [106]. Other possible therapeutical ways could be based on microbiota changes, since it has been suggested that the presence of some bacteria in the gut of aged people may contribute to AD progression [107, 108], affecting Aβ pathology. However, although Aβ accumulation is a factor for AD pathogenesis, in the absence of tau, its presence is insufficient for cognitive decline. Now, it has been described on microbiota-dependent progression of neurodegeneration in a mouse model of tauopathy, being that effect modulated by ApoE4 isoform [60]. In that way, reshaping or eliminating gut microbiota may decrease the progression of neurodegeneration.

    About new biomarkers, from the periphery, testing the level of formic acid in patients’ urine [81] or measuring the lower degree of pain, related to PNS, observed in subjects with mild cognitive decline, previous to dementia onset, could be suggested [32].

    Thus, to get a whole picture of the clinical features of a patient, at the early phases of AD continuum, the suggested Alzheimer’s precision medicine initiative could be a good approach to retard the progression of AD [109]. Nevertheless, more work should be done to look for modifiers of the disease that may prevent, decrease or stop the development of the disease.

    Abbreviations

    AD:

    Alzheimer’s disease

    ApoE4:

    apolipoprotein E4

    Aβ:

    beta amyloid

    CNS:

    cerebral nervous system

    CSF:

    cerebral spinal fluid

    FAD:

    familiar Alzheimer’s disease

    OA:

    osteoarthritis

    PNS:

    peripheral nervous system

    PSEN 1 :

    presenilin 1

    SAD:

    sporadic Alzheimer’s disease

    Declarations

    Acknowledgments

    We thank Ms. Nuria de la Torre Alonso for technical and editorial assistance. Figure 1 was created with Biorender (BioRender.com).

    Author contributions

    JA: Conceptualization, Investigation, Writing—original draft, Writing—review & editing. MP, MAV, ISM and FH: Conceptualization, Investigation. All authors read and approved the submitted version.

    Conflict of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    This work has been supported by grants from the Spanish Ministry of Science and Innovation [PID2020-113204GB-I00] (FH) and [PID2021-123859OB-100] from MCIN /AEI/10.13039/501100011033/FEDER, UE (JA). This work was supported by CSIC through an intramural grant [201920E104] (JA). The Centro de Biología Molecular Severo Ochoa (CBMSO) is a Severo Ochoa Center of Excellence (MICIN, award CEX2021-001154-S). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2023.

    References

    Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer’s disease. Nat Rev Dis Primers. 2015;1:15056. [DOI] [PubMed]
    Jensen AMG, Kitago Y, Fazeli E, Vaegter CB, Small SA, Petsko GA, et al. Dimerization of the Alzheimer’s disease pathogenic receptor SORLA regulates its association with retromer. Proc Natl Acad Sci U S A. 2023;120:e2212180120. [DOI] [PubMed]
    Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet. 2007;39:16877. [DOI] [PubMed] [PMC]
    Fernandez-Nogales M, Santos-Galindo M, Hernandez IH, Cabrera JR, Lucas JJ. Faulty splicing and cytoskeleton abnormalities in Huntington’s disease. Brain Pathol. 2016;26:7728. [DOI] [PubMed] [PMC]
    Goutagny R, Gu N, Cavanagh C, Jackson J, Chabot JG, Quirion R, et al. Alterations in hippocampal network oscillations and theta-gamma coupling arise before Aβ overproduction in a mouse model of Alzheimer’s disease. Eur J Neurosci. 2013;37:1896902. [DOI] [PubMed]
    Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:3536. [DOI] [PubMed]
    Du X, Wang X, Geng M. Alzheimer’s disease hypothesis and related therapies. Transl Neurodegener. 2018;7:2. [DOI] [PubMed] [PMC]
    Davies P. Challenging the cholinergic hypothesis in Alzheimer disease. JAMA. 1999;281:14334. [DOI] [PubMed]
    Khachaturian ZS. Calcium hypothesis of Alzheimer’s disease and brain aging. Ann N Y Acad Sci. 1994;747:111. [DOI] [PubMed]
    Zhu X, Raina AK, Perry G, Smith MA. Alzheimer’s disease: the two-hit hypothesis. Lancet Neurol. 2004;3:21926. [DOI] [PubMed]
    Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:15772. [DOI] [PubMed]
    Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci U S A. 1975;72:185862. [DOI] [PubMed] [PMC]
    Chen Z, Zhong C. Decoding Alzheimer’s disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol. 2013;108:2143. [DOI] [PubMed]
    Arnsten AFT, Datta D, Del Tredici K, Braak H. Hypothesis: tau pathology is an initiating factor in sporadic Alzheimer’s disease. Alzheimers Dement. 2021;17:11524. [DOI] [PubMed] [PMC]
    Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019;15:56581. [DOI] [PubMed]
    Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99118. [DOI] [PubMed] [PMC]
    Liang W, Chen Q, Cheng S, Wei R, Li Y, Yao C, et al. Skin chronological aging drives age-related bone loss via secretion of cystatin-A. Nature Aging. 2022;2:90622. [DOI] [PubMed]
    Gupta DP, Lee YS, Choe Y, Kim KT, Song GJ, Hwang SC. Knee osteoarthritis accelerates amyloid beta deposition and neurodegeneration in a mouse model of Alzheimer’s disease. Mol Brain. 2023;16:1. [DOI] [PubMed] [PMC]
    Eiser AR, Fulop T. Alzheimer’s disease is a multi-organ disorder: it may already be preventable. J Alzheimers Dis. 2023;91:127781. [DOI] [PubMed]
    Jorfi M, Maaser-Hecker A, Tanzi RE. The neuroimmune axis of Alzheimer’s disease. Genome Med. 2023;15:6. [DOI] [PubMed] [PMC]
    Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791800. [DOI] [PubMed]
    Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82:23959. [DOI] [PubMed]
    Avila J, Lucas JJ, Perez M, Hernandez F. Role of tau protein in both physiological and pathological conditions. Physiol Rev. 2004;84:36184. [DOI] [PubMed]
    Cha HJ, Shen J, Kang J. Regulation of gene expression by the APP family in the adult cerebral cortex. Sci Rep. 2022;12:66. [DOI] [PubMed] [PMC]
    Guo Q, Li H, Gaddam SS, Justice NJ, Robertson CS, Zheng H. Amyloid precursor protein revisited: neuron-specific expression and highly stable nature of soluble derivatives. J Biol Chem. 2012;287:243745. [DOI] [PubMed] [PMC]
    Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. J Cell Biol. 2018;217:45972. [DOI] [PubMed] [PMC]
    Paolicelli RC, Sierra A, Stevens B, Tremblay ME, Aguzzi A, Ajami B, et al. Microglia states and nomenclature: a field at its crossroads. Neuron. 2022;110:345883. [DOI] [PubMed] [PMC]
    Zhang G, Wang Z, Hu H, Zhao M, Sun L. Microglia in Alzheimer’s disease: a target for therapeutic intervention. Front Cell Neurosci. 2021;15:749587. [DOI] [PubMed] [PMC]
    Perea JR, Bolos M, Avila J. Microglia in Alzheimer’s disease in the context of tau pathology. Biomolecules. 2020;10:1439. [DOI] [PubMed] [PMC]
    Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med. 2016;213:66775. [DOI] [PubMed] [PMC]
    Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:4817. [DOI] [PubMed] [PMC]
    Qian X, Yue L, Mellor D, Robbins NM, Li W, Xiao S. Reduced peripheral nerve conduction velocity is associated with Alzheimer’s disease: a cross-sectional study from China. Neuropsychiatr Dis Treat. 2022;18:23142. [DOI] [PubMed] [PMC]
    Barthelemy NR, Li Y, Joseph-Mathurin N, Gordon BA, Hassenstab J, Benzinger TLS, et al. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat Med. 2020;26:398407. [DOI] [PubMed] [PMC]
    Blennow K, Dubois B, Fagan AM, Lewczuk P, de Leon MJ, Hampel H. Clinical utility of cerebrospinal fluid biomarkers in the diagnosis of early Alzheimer’s disease. Alzheimers Dement. 2015;11:5869. [DOI] [PubMed] [PMC]
    Avila J, Perry G. A multilevel view of the development of Alzheimer’s disease. Neuroscience. 2021;457:28393. [DOI] [PubMed]
    Ma YH, Shen LX, Li YZ, Leng Y, Yang L, Chen SD, et al. Lung function and risk of incident dementia: a prospective cohort study of 431,834 individuals. Brain Behav Immun. 2023;109:32130. [DOI] [PubMed]
    Tublin JM, Adelstein JM, Del Monte F, Combs CK, Wold LE. Getting to the heart of Alzheimer disease. Circ Res. 2019;124:1429. [DOI] [PubMed] [PMC]
    Peikert A, Cunningham JW. Amyloid-β and the risk of heart failure: cause or only association? JACC Heart Fail. 2023;11:1035. [DOI] [PubMed]
    Troncone L, Luciani M, Coggins M, Wilker EH, Ho CY, Codispoti KE, et al. Aβ amyloid pathology affects the hearts of patients with Alzheimer’s disease: mind the heart. J Am Coll Cardiol. 2016;68:2395407. [DOI] [PubMed] [PMC]
    Cardiogenic dementia. Lancet. 1977;309:278. [PubMed]
    SPRINT MIND Investigators for the SPRINT Research Group; Nasrallah IM, Pajewski NM, Auchus AP, Chelune G, Cheung AK, Cleveland ML, et al. Association of intensive vs. standard blood pressure control with cerebral white matter lesions. JAMA. 2019;322:52434. [DOI] [PubMed] [PMC]
    Moreno-Jimenez EP, Flor-Garcia M, Terreros-Roncal J, Rabano A, Cafini F, Pallas-Bazarra N, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25:55460. [DOI] [PubMed]
    Villeda SA, Luo J, Mosher KI, Zou B, Britschgi M, Bieri G, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477:904. [DOI] [PubMed] [PMC]
    Liu ZH, Wang YJ, Bu XL. Alzheimer’s disease: targeting the peripheral circulation. Mol Neurodegener. 2023;18:3. [DOI] [PubMed] [PMC]
    Bu XL, Xiang Y, Jin WS, Wang J, Shen LL, Huang ZL, et al. Blood-derived amyloid-β protein induces Alzheimer’s disease pathologies. Mol Psychiatry. 2018;23:194856. [DOI] [PubMed]
    Sun HL, Chen SH, Yu ZY, Cheng Y, Tian DY, Fan DY, et al. Blood cell-produced amyloid-β induces cerebral Alzheimer-type pathologies and behavioral deficits. Mol Psychiatry. 2021;26:556877. [DOI] [PubMed]
    Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019;25:4860. [DOI] [PubMed] [PMC]
    Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, Hyman B, et al. The Alzheimer’s disease-associated amyloid β-protein is an antimicrobial peptide. PLoS One. 2010;5:e9505. [DOI] [PubMed] [PMC]
    Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, et al. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 2016;8:340ra72. [DOI] [PubMed] [PMC]
    Chen C, Zhou Y, Wang H, Alam A, Kang SS, Ahn EH, et al. Gut inflammation triggers C/EBPβ/δ-secretase-dependent gut-to-brain propagation of Aβ and Tau fibrils in Alzheimer’s disease. EMBO J. 2021;40:e106320. [DOI] [PubMed] [PMC]
    Jin J, Xu Z, Zhang L, Zhang C, Zhao X, Mao Y, et al. Gut-derived β-amyloid: likely a centerpiece of the gut-brain axis contributing to Alzheimer’s pathogenesis. Gut Microbes. 2023;15:2167172. [DOI] [PubMed] [PMC]
    Sun Y, Sommerville NR, Liu JYH, Ngan MP, Poon D, Ponomarev ED, et al. Intra-gastrointestinal amyloid-β1-42 oligomers perturb enteric function and induce Alzheimer’s disease pathology. J Physiol. 2020;598:420923. [DOI] [PubMed] [PMC]
    Carloni S, Bertocchi A, Mancinelli S, Bellini M, Erreni M, Borreca A, et al. Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science. 2021;374:43948. [DOI] [PubMed]
    Dai CL, Liu F, Iqbal K, Gong CX. Gut microbiota and immunotherapy for Alzheimer’s disease. Int J Mol Sci. 2022;23:15230. [DOI] [PubMed] [PMC]
    Mawe GM, Hoffman JM. Serotonin signalling in the gut—functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2013;10:47386. [DOI] [PubMed] [PMC]
    Avila-Villanueva M, Gomez-Ramirez J, Avila J, Fernandez-Blazquez MA. Alzheimer’s disease and empathic abilities: the proposed role of the cingulate cortex. J Alzheimers Dis Rep. 2021;5:34552. [DOI] [PubMed] [PMC]
    Canton-Habas V, Rich-Ruiz M, Romero-Saldana M, Carrera-Gonzalez MDP. Depression as a risk factor for dementia and Alzheimer’s disease. Biomedicines. 2020;8:457. [DOI] [PubMed] [PMC]
    Konopelski P, Mogilnicka I. Biological effects of indole-3-propionic acid, a gut microbiota-derived metabolite, and its precursor tryptophan in mammals’ health and disease. Int J Mol Sci. 2022;23:1222. [DOI] [PubMed] [PMC]
    Gheorghe CE, Martin JA, Manriquez FV, Dinan TG, Cryan JF, Clarke G. Focus on the essentials: tryptophan metabolism and the microbiome-gut-brain axis. Curr Opin Pharmacol. 2019;48:13745. [DOI] [PubMed]
    Seo DO, O’Donnell D, Jain N, Ulrich JD, Herz J, Li Y, et al. ApoE isoform– and microbiota-dependent progression of neurodegeneration in a mouse model of tauopathy. Science. 2023;379:eadd1236. [DOI] [PubMed]
    Agirman G, Hsiao EY. Gut microbes shape athletic motivation. Nature. 2022;612:6334. [DOI] [PubMed]
    Goncalves RA, Sudo FK, Lourenco MV, Drummond C, Assuncao N, Vanderborght B, et al. Cerebrospinal fluid irisin and lipoxin A4 are reduced in elderly Brazilian individuals with depression: insight into shared mechanisms between depression and dementia. Alzheimers Dement. 2023;19:2595604. [DOI] [PubMed]
    Peng J, Wu J. Effects of the FNDC5/irisin on elderly dementia and cognitive impairment. Front Aging Neurosci. 2022;14:863901. [DOI] [PubMed] [PMC]
    Bassendine MF, Taylor-Robinson SD, Fertleman M, Khan M, Neely D. Is Alzheimer’s disease a liver disease of the brain? J Alzheimers Dis. 2020;75:114. [DOI] [PubMed] [PMC]
    Liu YH, Wang YR, Xiang Y, Zhou HD, Giunta B, Manucat-Tan NB, et al. Clearance of amyloid-beta in Alzheimer’s disease: shifting the action site from center to periphery. Mol Neurobiol. 2015;51:17. [DOI] [PubMed]
    Xiang Y, Bu XL, Liu YH, Zhu C, Shen LL, Jiao SS, et al. Physiological amyloid-beta clearance in the periphery and its therapeutic potential for Alzheimer’s disease. Acta Neuropathol. 2015;130:48799. [DOI] [PubMed] [PMC]
    Cheng Y, He CY, Tian DY, Chen SH, Ren JR, Sun HL, et al. Physiological β-amyloid clearance by the liver and its therapeutic potential for Alzheimer’s disease. Acta Neuropathol. 2023;145:71731. [DOI] [PubMed]
    Nho K, Kueider-Paisley A, Ahmad S, MahmoudianDehkordi S, Arnold M, Risacher SL, et al.; Alzheimer’s Disease Neuroimaging Initiative and the Alzheimer Disease Metabolomics Consortium. Association of altered liver enzymes with Alzheimer disease diagnosis, cognition, neuroimaging measures, and cerebrospinal fluid biomarkers. JAMA Netw Open. 2019;2:e197978. [DOI] [PubMed] [PMC]
    Giannisis A, Patra K, Edlund AK, Nieto LA, Benedicto-Gras J, Moussaud S, et al. Brain integrity is altered by hepatic APOE ε4 in humanized-liver mice. Mol Psychiatry. 2022;27:353343. [DOI] [PubMed] [PMC]
    Liu CC, Zhao J, Fu Y, Inoue Y, Ren Y, Chen Y, et al. Peripheral apoE4 enhances Alzheimer’s pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci. 2022;25:102033. [DOI] [PubMed] [PMC]
    Lam V, Takechi R, Hackett MJ, Francis R, Bynevelt M, Celliers LM, et al. Synthesis of human amyloid restricted to liver results in an Alzheimer disease-like neurodegenerative phenotype. PLoS Biol. 2021;19:e3001358. [DOI] [PubMed] [PMC]
    Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, Franceschi C, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med. 2019;25:182232. [DOI] [PubMed] [PMC]
    Xue YH, Peng YS, Ting HF, Peijer Hsieh J, Huang YK, Wang YH, et al. Etoricoxib and diclofenac might reduce the risk of dementia in patients with osteoarthritis: a nation-wide, population-based retrospective cohort study. Dement Geriatr Cogn Disord. 2018;45:26271. [DOI] [PubMed]
    Tian DY, Cheng Y, Zhuang ZQ, He CY, Pan QG, Tang MZ, et al. Physiological clearance of amyloid-beta by the kidney and its therapeutic potential for Alzheimer’s disease. Mol Psychiatry. 2021;26:607482. [DOI] [PubMed]
    Montejo de Garcini E, Diez JC, Avila J. Quantitation and characterization of tau factor in porcine tissues. Biochim Biophys Acta. 1986;881:45661. [DOI] [PubMed]
    Valles-Saiz L, Peinado-Cahuchola R, Avila J, Hernandez F. Microtubule-associated protein tau in murine kidney: role in podocyte architecture. Cell Mol Life Sci. 2022;79:97. [DOI] [PubMed] [PMC]
    Doran M, du Plessis DG, Ghadiali EJ, Mann DM, Pickering-Brown S, Larner AJ. Familial early-onset dementia with tau intron 10 + 16 mutation with clinical features similar to those of Alzheimer disease. Arch Neurol. 2007;64:15359. [DOI] [PubMed]
    Stocker H, Beyer L, Trares K, Perna L, Rujescu D, Holleczek B, et al. Association of kidney function with development of Alzheimer disease and other dementias and dementia-related blood biomarkers. JAMA Netw Open. 2023;6:e2252387. [DOI] [PubMed]
    Wang S, Wang J, Dove A, Guo J, Yang W, Qi X, et al. Association of impaired kidney function with dementia and brain pathologies: a community-based cohort study. Alzheimers Dement. 2022;[Epub ahead of print]. [DOI] [PubMed]
    Xu H, Garcia-Ptacek S, Bruchfeld A, Fu EL, Shori TD, Lindholm B, et al. Association between cholinesterase inhibitors and kidney function decline in patients with Alzheimer’s dementia. Kidney Int. 2023;103:16676. [DOI] [PubMed]
    Chen F, Wang N, Tian X, Qin Y, Su J, He R, et al. The potential diagnostic accuracy of urine formaldehyde levels in Alzheimer’s disease: a systematic review and meta-analysis. Front Aging Neurosci. 2022;14:1057059. [DOI] [PubMed] [PMC]
    Mathieu C, Martens PJ, Vangoitsenhoven R. One hundred years of insulin therapy. Nat Rev Endocrinol. 2021;17:71525. [DOI] [PubMed]
    Neumann KF, Rojo L, Navarrete LP, Farias G, Reyes P, Maccioni RB. Insulin resistance and Alzheimer’s disease: molecular links & clinical implications. Curr Alzheimer Res. 2008;5:43847. [DOI] [PubMed]
    Li L, Holscher C. Common pathological processes in Alzheimer disease and type 2 diabetes: a review. Brain Res Rev. 2007;56:384402. [DOI] [PubMed]
    Desai GS, Zheng C, Geetha T, Mathews ST, White BD, Huggins KW, et al. The pancreas-brain axis: insight into disrupted mechanisms associating type 2 diabetes and Alzheimer’s disease. J Alzheimers Dis. 2014;42:34756. [DOI] [PubMed]
    de la Monte SM, Wands JR. Alzheimer’s disease is type 3 diabetes-evidence reviewed. J Diabetes Sci Technol. 2008;2:110113. [DOI] [PubMed] [PMC]
    Reish NJ, Jamshidi P, Stamm B, Flanagan ME, Sugg E, Tang M, et al. Multiple cerebral hemorrhages in a patient receiving lecanemab and treated with t-PA for stroke. N Engl J Med. 2023;388:4789. [DOI] [PubMed] [PMC]
    Alves F, Kallinowski P, Ayton S. Accelerated brain volume loss caused by anti-beta-amyloid drugs: a systematic review and meta-analysis. Neurology. 2023;100:e211424. [DOI] [PubMed]
    Moutinho S. The long road to a cure for Alzheimer’s disease is paved with failures. Nat Med. 2022;28:222831. [DOI] [PubMed]
    Cummings J, Lee G, Nahed P, Kambar M, Zhong K, Fonseca J, et al. Alzheimer’s disease drug development pipeline: 2022. Alzheimers Dement (N Y). 2022;8:e12295. [DOI] [PubMed] [PMC]
    Aguillon D, Langella S, Chen Y, Sanchez JS, Su Y, Vila-Castelar C, et al. Plasma p-tau217 predicts in vivo brain pathology and cognition in autosomal dominant Alzheimer’s disease. Alzheimers Dement. 2023;19:258594. [DOI] [PubMed]
    Zhao A, Jiao Y, Ye G, Kang W, Tan L, Li Y, et al. Soluble TREM2 levels associate with conversion from mild cognitive impairment to Alzheimer’s disease. J Clin Invest. 2022;132:e158708. [DOI] [PubMed] [PMC]
    Franco R, Cedazo-Minguez A. Successful therapies for Alzheimer’s disease: why so many in animal models and none in humans? Front Pharmacol. 2014;5:146. [DOI] [PubMed] [PMC]
    Perez-Olives C, Rivas-Santisteban R, Lillo J, Navarro G, Franco R. Recent advances in the potential of cannabinoids for neuroprotection in Alzheimer’s, Parkinson’s, and Huntington’s diseases. Adv Exp Med Biol. 2021;1264:8192. [DOI] [PubMed]
    Levert S, Pilliod J, Aumont E, Armanville S, Tremblay C, Calon F, et al. Direct and indirect effects of filamin A on tau pathology in neuronal cells. Mol Neurobiol. 2023;60:102139. [DOI] [PubMed] [PMC]
    Else H. Alzheimer’s drug saga prompts journal to scrutinize whistle-blowers. Nature. 2023;613:6189. [DOI] [PubMed]
    Yu H, Liu Y, He B, He T, Chen C, He J, et al. Platelet biomarkers for a descending cognitive function: a proteomic approach. Aging Cell. 2021;20:e13358. [DOI] [PubMed] [PMC]
    Huang YM, Ma YH, Gao PY, Wang ZB, Huang LY, Hou JH, et al. Plasma β2-microglobulin and cerebrospinal fluid biomarkers of Alzheimer’s disease pathology in cognitively intact older adults: the CABLE study. Alzheimers Res Ther. 2023;15:69. [DOI] [PubMed] [PMC]
    Bu XL, Sun PY, Fan DY, Wang J, Sun HL, Cheng Y, et al. Associations of plasma soluble CD22 levels with brain amyloid burden and cognitive decline in Alzheimer’s disease. Sci Adv. 2022;8:eabm5667. [DOI] [PubMed]
    Avila-Villanueva M, Gomez-Ramirez J, Maestu F, Venero C, Avila J, Fernandez-Blazquez MA. The role of chronic stress as a trigger for the Alzheimer disease continuum. Front Aging Neurosci. 2020;12:561504. [DOI] [PubMed] [PMC]
    Rocca WA, Petersen RC, Knopman DS, Hebert LE, Evans DA, Hall KS, et al. Trends in the incidence and prevalence of Alzheimer’s disease, dementia, and cognitive impairment in the United States. Alzheimers Dement. 2011;7:8093. [DOI] [PubMed] [PMC]
    Wolters FJ, Chibnik LB, Waziry R, Anderson R, Berr C, Beiser A, et al. Twenty-seven-year time trends in dementia incidence in Europe and the United States: The Alzheimer Cohorts Consortium. Neurology. 2020;95:e51931. [DOI] [PubMed] [PMC]
    Morris MC, Tangney CC, Wang Y, Sacks FM, Bennett DA, Aggarwal NT. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimers Dement. 2015;11:100714. [DOI] [PubMed] [PMC]
    Rodriguez-Matellan A, Alcazar N, Hernandez F, Serrano M, Avila J. In vivo reprogramming ameliorates aging features in dentate gyrus cells and improves memory in mice. Stem Cell Reports. 2020;15:105666. [DOI] [PubMed] [PMC]
    Han X, Zhang T, Liu H, Mi Y, Gou X. Astrocyte senescence and Alzheimer’s disease: a review. Front Aging Neurosci. 2020;12:148. [DOI] [PubMed] [PMC]
    Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288:51836. [DOI] [PubMed] [PMC]
    Jiang C, Li G, Huang P, Liu Z, Zhao B. The gut microbiota and Alzheimer’s disease. J Alzheimers Dis. 2017;58:115. [DOI] [PubMed]
    Varesi A, Pierella E, Romeo M, Piccini GB, Alfano C, Bjørklund G, et al. The potential role of gut microbiota in Alzheimer’s disease: from diagnosis to treatment. Nutrients. 2022;14:668. [DOI] [PubMed] [PMC]
    Hampel H, Vergallo A, Perry G, Lista S; Alzheimer Precision Medicine Initiative (APMI). The Alzheimer Precision Medicine Initiative. J Alzheimers Dis. 2019;68:124. [DOI] [PubMed] [PMC]