Skip to main content

Peripheral proteinopathy in neurodegenerative diseases

Abstract

Proteinopathies in neurology typically refer to pathological changes in proteins associated with neurological diseases, such as the aggregation of amyloid β and Tau in Alzheimer’s disease, α-synuclein in Parkinson’s disease and multiple system atrophy, and TAR DNA-binding protein 43 in amyotrophic lateral sclerosis and frontotemporal dementia. Interestingly, these proteins are also commonly found in peripheral tissues, raising important questions about their roles in neurological disorders. Multiple studies have shown that peripherally derived pathological proteins not only travel to the brain through various routes, aggravating brain pathology, but also contribute significantly to peripheral dysfunction, highlighting their crucial impact on neurological diseases. Investigating how these peripherally derived proteins influence the progression of neurological disorders could open new horizons for achieving early diagnosis and treatment. This review summarizes the distribution, transportation pathways, and pathogenic mechanisms of several neurodegenerative disease-related pathological proteins in the periphery, proposing that targeting these peripheral pathological proteins could be a promising strategy for preventing and managing neurological diseases.

Introduction

Neurodegenerative diseases encompass a range of conditions impacting the central nervous system (CNS), including Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple system atrophy (MSA), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and several other diseases. These diseases typically result from the abnormal aggregation of proteins in the CNS, such as amyloid-β (Aβ) plaques and tubulin-associated unit (Tau) for AD, α-synuclein (α-syn) in neurons for PD, α-syn in oligodendrocytes for MSA, TAR DNA-binding protein 43 (TDP-43) for ALS and a type of FTD, leading to gradual deterioration of neural structure and function [1,2,3,4]. With the increasing aging population globally, the incidence of these diseases is escalating annually, presenting serious challenges to global public health [5, 6]. Current therapies primarily focus on managing symptoms to improve the quality of life for patients [7, 8]. However, achieving a fundamental cure remains challenging. Therefore, it is urgent and imperative to explore the deeper mechanisms underlying neurodegenerative diseases and to develop corresponding treatments targeting these mechanisms.

While traditional research has primarily focused on the pathological proteins in the CNS [9], increasing evidence from human proteomics research indicates that these proteins are also expressed in various peripheral organs [10,11,12]. Recently, increasing studies have demonstrated that peripheral tissues, such as the gastrointestinal tract, express pathological proteins at elevated levels in patients with neurodegenerative disease, highlighting their potential role in driving disease progression [13,14,15]. The peripheral pathological proteins have been confirmed to not only be transported to the brain, inducing CNS pathology, but also to play a role in regulating homeostasis within the peripheral system.

This review summarizes the peripheral distribution and physiological functions of Aβ, Tau, α-syn, and TDP-43, the potential mechanisms of their transportation into the brain, and their roles in the pathogenesis of neurodegenerative diseases. In addition, we discuss therapeutic approaches targeting peripheral pathological proteins in neurodegenerative diseases. Our review contributes to comprehending the relationship between peripheral proteinopathies and neurodegenerative diseases, providing novel insights and strategies for their prevention and treatment.

Origins and functions of peripheral key proteins

Distribution and function of peripheral Aβ

Aβ was initially characterized by Dr. Glenner and Dr. Wong from the brain blood vessels of AD patients [16], and several subsequent studies have affirmed that Aβ is one of the key pathological features of AD [17, 18]. While Aβ is primarily produced in the brain, studies have found that a wide range of peripheral tissues such as the liver, kidney, spleen, intestine, salivary gland, adrenal gland, heart, pancreas, thyroid gland, endothelial lymph nodes, retina, skeletal muscle, skin and various blood cells also extensively express the amyloid precursor protein (APP) gene, with corresponding production of peripheral APP and related peptides (Fig. 1, Table 1) [19,20,21]. In the skeletal muscle, although the amount of Aβ produced by skeletal muscle cells is relatively small, nearly one-third of the total body weight of skeletal muscle still expresses high levels of Aβ in peripheral tissues [22, 23]. In human peripheral blood, cells can internalize APP and produce peripheral Aβ, with platelets being the major source, accounting for over 90% of Aβ [24, 25]. Differential expression of Aβ in the skin has been observed in AD patients, and Aβ aggregates may form in the skin or subcutaneous tissue [26]. The liver and kidney are the main organs responsible for Aβ metabolism and clearance in the body, while their hepatocytes and renal tubular epithelial cells can also produce APP and break it down into Aβ [27, 28]. The intestine is another important organ with high expression of peripheral Aβ. Studies have reported elevated intestinal Aβ aggregates in AD patients, possibly contributing to the pathological progression of AD [29]. It should be noted that while multiple peripheral tissues can express the APP gene and produce Aβ, the predominant form of Aβ generated by peripheral tissue proteolysis is Aβ40, which is different from the more neurotoxic Aβ42 subtype synthesized primarily by CNS cells [30,31,32,33], with much less Aβ aggregation in peripheral tissues at least in physiological conditions. The differential extent of Aβ aggregation between the peripheral organs and brain may also be attributed to the different isoforms of APP in the periphery versus the CNS [33], ranging in size from 695 to 770 amino acids. The most abundant form in the brain (APP695) is produced mainly by neurons and differs from longer forms of APP expressed by peripheral cells and platelets (APP751 and APP770 isoforms) in that the CNS form lacks a Kunitz-type protease inhibitor sequence in its ectodomain [34, 35].

Fig. 1
figure 1

The distribution of neurodegenerative disease-related pathological proteins in peripheral tissues. The characteristic pathological proteins of neurodegenerative diseases, including Aβ, Tau, α-syn and TDP-43, are not only expressed in the CNS but are also widely present in peripheral tissues such as blood, gastrointestinal tract, liver, kidney, lung, heart, skin, muscles, bone, and peripheral nerves. The levels of these pathological proteins in the periphery are regulated by the peripheral environment including bacteria and inflammation. These proteins perform various physiological and pathological functions in peripheral tissues

Table 1 Summary of recent studies on protein properties in central and peripheral systems

Despite the difference in isoforms, Joachim et al. have reported that Aβ deposits are detected in non-neural tissues, including skin, subcutaneous tissue, intestine and blood vessels, of AD patients and elderly normal subjects [36]. Similarly, aggregated Aβ has been found in the impaired myocardial tissue and retina of AD patients [37, 38]. Additionally, amyloid-like deposits accumulate during natural aging in the body wall muscle of C. elegans, in human primary myotubes, and in mouse skeletal muscle [39]. The support for Aβ aggregation can also be found in the reports that β- and γ-secretases, two enzymes essential for the generation of the toxic Aβ species [40, 41], are present in peripheral organs. Specifically, β-secretase (BACE1) mRNA and protein are found in many cell types including pancreatic β-cells, adipocytes, hepatocytes, and vascular cells [42]. γ-Secretase is a multiprotein complex consisting of presenilin, nicastrin, anterior-pharynx defective-1, and presenilin enhancer-2 [43]. γ-Secretase is clearly detected in the spleen, kidneys, lungs, pancreas, and liver, although, like BACE1, its expression level is much higher in the brain and the spinal cord [44].

While Aβ and its aggregates are found in various peripheral tissues and cells of the human body, the physiological functions of peripheral Aβ have not been fully elucidated. Research has discovered the antimicrobial activity of Aβ by inhibiting the growth and proliferation of bacteria in gut [45, 46]. Conversely, bacteria can also influence the expression and aggregation of Aβ [47]. Transplanting fecal microbiota from APP/PS1 mice into wild-type mice significantly increases intestinal Aβ expression, exacerbating neuroinflammation and Aβ plaques in the CNS. In contrast, transplantation of fecal microbiota from healthy wild-type mice ameliorates AD pathological changes in ADLPAPT transgenic mice [48]. Numerous studies have reported that the dysbiosis of gut microbiota in AD patients leads to abnormal elevation of Aβ expression in the intestine [49,50,51]. Additionally, Aβ has potential anti-cancer effects [52] although the underlying mechanisms are not fully understood. Studies have shown that Aβ can inhibit the growth of human glioblastoma and adenocarcinoma tumors by regulating the cell cycle, suppressing tumor angiogenesis, inducing apoptosis of tumor cells and reducing the blood supply needed for tumor growth [53].

The immunomodulatory effects of peripheral Aβ are controversial and not yet fully confirmed. Some studies have shown that Aβ can influence the activity and function of macrophages in APP/PS1 mice, regulating the release of inflammatory factors and affecting the body’s inflammatory response [54, 55]. Li et al. found that Aβ exerts a biphasic effect at different stages of AD, suppressing peripheral inflammatory macrophages in the early stage of the disease while promoting them in the late stage [54, 55]. Aβ also affects the function of T cells, B cells, and other immune cells in human immune system, including cell proliferation, differentiation, and activation, thereby influencing the immune response [56, 57]. However, there is still significant debate on the exact mechanisms of immune regulation by Aβ and the impact of peripheral Aβ on brain immunity and AD pathogenesis (see below for more detailed discussion).

Distribution and function of peripheral Tau

Tau, first discovered by Dr. Kirschner’s laboratory in 1975 [58], is a microtubule-associated protein primarily expressed in neurons, where it plays a role in microtubule stabilization and axonal transport [59]. The CNS Tau is encoded by 6-kb mRNA and exists as six isoforms with molecular weights ranging from 48 to 67 kDa. In contrast, peripheral Tau, often referred to as ‘big Tau’, is a larger high-molecular-weight isoform (approximately 110 kDa) produced from an alternative splicing of the Tau mRNA transcript, resulting in an 8-kb mRNA variant. All of these isoforms are encoded by the same gene [60]. In the peripheral ganglia of neonatal animals, both 6-kb and 8-kb tau mRNAs are present, whereas in adult animals, only the 8-kb mRNA is detected peripherally [61]. This indicates that Tau mRNA undergoes different splicing in the central and peripheral nervous systems of vertebrates, with the big Tau being specifically expressed in the peripheral nervous system [62]. Tau is found in many peripheral tissues such as the peripheral nervous system, lungs, heart, intestines, gallbladders, submandibular glands, kidneys, bone, muscles, skin, and retina (Fig. 1, Table 1) [63,64,65,66,67,68,69]. Peripheral nerves, including those in the heart and intestines, are the primary sources of peripheral Tau [70]. In addition to peripheral neurons, cardiomyocytes in the heart, epithelial cells in the lungs, and enteroendocrine cells in the intestines can also produce Tau [12, 64, 71].

Peripheral big Tau undergoes post-translational modifications, such as phosphorylation and acetylation [72]. Dysregulation of these processes may contribute to the formation of hyperphosphorylated and misfolded Tau, which can then enter the circulation and potentially the CNS [73]. To this end, abnormal phosphorylation and aggregation of CNS Tau is a critical pathological mechanism in AD [74, 75]. However, it is more difficult for peripheral big Tau to form aggregates, due to its complex structural characteristics compared to the CNS Tau [76, 77]. The precise impact of peripheral big Tau on CNS Tau in neurodegeneration remains to be fully understood. Additionally, the influence of big Tau species on the detection of phosphorylated peripheral Tau as a biomarker requires further investigation. For example, Tau hyperphosphorylation at Ser396 has been detected in the hearts of patients with heart failure and AD, which may lead to formation of toxic oligomers [78, 79].

In peripheral tissues, Tau may have functions beyond microtubule stabilization. For instance, the expression of Tau increases in pulmonary endothelial cells during bacterial pneumonia and sepsis [80, 81]. Although the specific pathological mechanisms are unclear, a series of studies have demonstrated that abnormalities in Tau can lead to skeletal muscle dysfunction and result in muscle weakness [82]. Targeting Tau through immunotherapy improves myocardial function, indicating that the pathological aggregation of Tau disrupts cardiac function [78, 79].

Distribution and function of peripheral α-syn

Traditionally, α-syn is believed to be a relatively small, soluble protein primarily expressed in the CNS, where it is involved in synaptic vesicle trafficking and neurotransmitter release [83]. The aggregation of α-syn-containing Lewy bodies in neurons or oligodendrocytes is a major pathological feature of PD or MSA [84, 85]. Though first discovered in the brain by Dr. Maroteaux, the protein is widely distributed in various tissues throughout the body (Fig. 1, Table 1) [86,87,88]. Studies have shown that α-syn is abundantly expressed in the intestine, especially in enteric neurons [89], and regulates gastrointestinal motility and secretion [90]. Its expression has also been detected in the heart, indicating its potential involvement in cardiovascular regulation [91]. Although SNCA mRNA is not expressed in the liver, α-syn protein accumulates there and serves as a major pathway for α-syn metabolism [92]. Additionally, over 99% of α-syn in the blood is carried by erythrocytes, making them an important source of peripheral α-syn [93]. It should be noted that due to structural differences between α-syn in peripheral red blood cells and the CNS, erythrocytic α-syn, which predominantly exists as stable, helically folded tetramers, is less prone to aggregation compared to brain α-syn [94, 95].

The physiological mechanisms of α-syn in the periphery have not been fully elucidated. α-Syn has physiological effects on the skin, regulating the proliferation and differentiation of skin cells and maintaining the skin barrier and skin nerve [96, 97]. Additionally, α-syn participates in immune regulation in the skin, modulating skin inflammation and immune responses [98]. The widespread expression of α-syn in the periphery is important evidence for the peripheral origin hypothesis of PD. Several factors such as inflammation, oxidative stress, and aging can promote the misfolding and aggregation of α-syn in peripheral tissues, which may then spread to the CNS via various routes [99]. Intestinal α-syn also causes gastrointestinal dysfunction in patients with PD [100]. Yang et al. found that α-syn can spontaneously aggregate in the intestine [101]. Challis et al. demonstrated that injection of α-syn fibrils into the gut can cause gastrointestinal impairment [102]. Further investigations are required to clarify whether peripheral aggregation of α-syn in other organs causes tissue damage or can be transported to the CNS.

Distribution and function of peripheral TDP-43

TDP-43, encoded by the TARDBP gene [103], was initially discovered in 1995 as a suppressor of HIV-1 gene expression. It is a highly conserved RNA/DNA-binding protein expressed throughout the body (Fig. 1, Table 1). In physiological conditions, TDP-43 is predominantly localized in the nucleus. However, it can shuttle between the nucleus and cytoplasm through active and passive transport, exerting physiological functions in the cytoplasm, including the regulation of mRNA transport, RNA metabolism, microRNA maturation, and stress granule formation [104]. TDP-43 is also found in mitochondria, where it interacts with the mitochondrial genome and plays a crucial role in respiratory chain pathways [105]. TDP-43 is essential for early embryogenesis as knockout of TARDBP leads to embryonic lethality in mice [106]. The mislocalization and aggregation of TDP-43 are considered key pathologies underlying neurodegenerative diseases including ALS and FTD [107]. Although no studies have yet reported whether there are differences between the central and peripheral isoforms of TDP-43, an increasing number of studies have begun to focus on the abnormal distribution and expression of TDP-43 in peripheral tissues (Table 1). TDP-43 aggregates are found in skeletal muscles of patients with neuromuscular diseases such as sporadic inclusion body myositis, familial ALS, and polymyositis [108, 109]. Recent autopsy studies have identified phosphorylated TDP-43 (pTDP-43, pSer409/410) aggregates in the skeletal and cardiac muscles as potential biomarkers for both ALS and non-ALS, such as sporadic inclusion body myositis, myofibrillar myopathies, hereditary motor and sensory neuropathy, etc [110, 111]. Further investigation indicates that pTDP-43 pathology is more commonly observed in axial muscles, i.e., the paraspinal muscles and diaphragm, and less frequently in appendicular muscles [112]. Moreover, pTDP-43 aggregation is also present in the intramuscular nerve bundle axons of ALS patients, suggesting that pTDP-43 in the intramuscular nerve bundles may be a peripheral pathological feature of ALS [113]. Despite these exciting discoveries, our understanding of the peripheral TDP-43 in normal and diseased conditions, such as ALS and FTD, is rather limited.

Transportation of peripheral key proteins to the brain

Before considering the influence of peripheral proteinopathies on the CNS, it is essential to understand the routes by which they are transported into the brain. All peripheral proteins discussed above can exhibit prion-like biophysical and biochemical properties [114, 115]. Unlike conventional viruses, prions lack genetic material (such as DNA or RNA) and propagate by inducing misfolding of normal proteins into an abnormal conformation, a process known as "seeding" [116, 117]. Many misfolded gut-derived proteins, including Aβ, Tau, and α-syn, have been shown to transmit to the brain via the vagal nerve, leading to neuronal death (Fig. 2) [118,119,120,121,122], through a prion-like mechanism. Additionally, these peripherally derived proteins can enter the brain directly via transporters on the blood–brain barrier (BBB) or through extracellular vesicles (EVs) secreted by peripheral tissue cells, especially when the BBB is compromised (Fig. 2) [123, 124] (see below for more detailed discussion). Much less is known about TDP-43 transmission. Finally, through transportation via the gut-brain axis, Aβ, Tau, α-syn, and TDP-43 derived from dietary sources or the gut microbiota may play a role in the pathogenesis of neurodegenerative diseases.

Fig. 2
figure 2

Transportation of peripheral pathological proteins into the brain. Peripheral pathological proteins can enter the brain through various pathways. Aβ, Tau, and α-syn enriched in the gastrointestinal tract can be transported via the vagus nerve. On the other hand, these pathological proteins can flow through the bloodstream from the periphery into the CNS. When the BBB remains intact, these proteins can cross into the CNS through specific transporters, such as AGER, which selectively transports Aβ. Additionally, EVs carrying these pathological proteins can be endocytosed by brain microvascular endothelial cells, allowing them to cross the BBB. When the BBB is compromised, these pathological proteins, whether in free form or encapsulated within EVs, can infiltrate the brain through the intercellular spaces

Vagus nerve

Peripheral Aβ, Tau, and α-syn, especially those derived from the intestine, can enter the brain through the vagus nerve, which is an important pathway in the pathogenesis of neurodegenerative diseases [121, 122, 125]. The vagus nerve, one of the longest peripheral nerves, originates from the brainstem and extends to the abdomen and pelvis, controlling the function of multiple visceral organs [126]. It can serve as a conduit for the spread of misfolded proteins from the periphery to the CNS. Several studies have found that intestinal injection of Aβ, Tau, and α-syn can promote the transportation of pathological proteins from the peripheral system to the CNS via the vagus nerve. Notably, severing the vagus nerve significantly inhibits the entry of these injected proteins into the brain [127,128,129]. We have also shown that gut Aβ can lead to neuronal damage, which can be alleviated by vagotomy, although the process seems to be age-dependent [130]. In addition to exogenously injecting these pathological proteins, Xiang et al. using newly developed gut-inducible SYN103+/- and TAU368+/- transgenic mouse models, further demonstrated that endogenous gut-derived α-syn and Tau can spontaneously aggregate and propagate to the brain via the vagus nerve [131]. However, it should be noted that some have suggested that vagal nerve transmission may not be significant in non-human primates [132], which could be attributed to factors such as the animal model used, the type of inoculum, the time after injection, and species differences.

BBB and transporter

Besides the vagus nerve, soluble pathological proteins in the circulatory system can directly interact with the transporter on the recipient cells. BBB plays a critical role in regulating the exchange of molecules between the peripheral circulation and the CNS [133]. Under normal conditions, the intact BBB effectively prevents the transport of large peripheral molecules into the brain. However, in pathological conditions such as aging, trauma, and inflammation, peripheral pathological proteins including Aβ, Tau, and α-syn can enter the brain through enhanced active transcytosis, including receptor-mediated and absorptive transcytosis, as well as passive diffusion through damaged BBB [134]. The increased transcytosis may be attributed to the upregulation of receptors, as recent research revealed that elevated expression of AGER, which encodes the receptor for advanced glycation end-products, in endothelial cells, promotes the transport of peripheral Aβ to the brain [135]. Specific transporters for other peripheral pathological proteins into the brain have not yet been identified. Further research is needed to uncover more transporters and their roles in the pathological progression of neurodegenerative diseases.

EVs

EVs are membrane-bound small vesicles produced by cells, playing a crucial role in intercellular communication and signaling by containing substances such as proteins, nucleic acids, and lipids [136]. In recent years, EVs have received increasing attention due to their ability to traverse the intact BBB. For example, erythrocyte-derived EVs carrying α-syn enter brain through the BBB [137,138,139]. By radioactively labeling red blood cell-derived EVs (RBC-EVs) with Na125I and DiI labeling, Matsumoto et al. found that these EVs can enter the brain even when the BBB is intact and are primarily phagocytosed by microglia [139]. Subsequently, in vivo imaging was conducted to track DiR-labeled RBC-EVs injected via the tail vein and radioactive 125I-labeled RBC-EVs injected into the gut, further confirming that the peripheral EVs can transfer to the brain [101, 140]. Immunostaining for DiI revealed that these RBC-EVs were captured by neurons and astrocytic end-feet [101, 140]. These studies also found that BBB disruption by lipopolysaccharide accelerates the entry of peripheral RBC-EVs into the brain [101, 139, 140]. These findings suggest that EVs may be one of the important pathways for peripheral proteins to cross the BBB and enter brain tissue, which is significant for understanding the pathogenesis of neurodegenerative diseases and identifying new therapeutic targets. However, the entry of peripheral EVs carrying Aβ, Tau and TDP-43 into the brain has not yet been confirmed. Further studies are required to validate the potential of other pathological proteins to enter the brain via EVs, as well as their exact roles and potential applications in neurodegenerative diseases.

Neuropathology

AD

Several studies have reported that peripheral Aβ and Tau play significant pathogenic roles in AD, with particular emphasis on Aβ produced in the gut, which is regulated by the gut microbiota. Gut-derived Aβ can enter the brain through various pathways and is believed to be an important pathogenic factor in the progression of AD.

Once transported to the CNS, peripherally derived Aβ and Tau may impair brain function through several pathways. First, Aβ exacerbates the formation and deposition of Aβ fibrils in the brain, which may be one of the main sources of Aβ plaques [13, 130]. Similarly, peripheral Tau can cross into the CNS and contribute to the spread of neurofibrillary tangles [127, 131]. Second, the aggregates of peripheral Aβ and Tau exert certain neurotoxicity and can directly induce neuronal death. Furthermore, gut-derived Aβ and Tau can trigger neuroinflammation, activating microglia, thereby promoting neuronal damage and affecting functions of the CNS [130, 141]. Finally, neuroinflammation induced by intra-gastrointestinal Aβ and Tau can further damage the BBB with a vicious cycle, allowing more peripheral Aβ to enter the brain tissue [120]. In summary, peripherally derived Aβ may negatively affect the CNS through multiple pathways, leading to the progression of AD neuropathology (Fig. 3).

Fig. 3
figure 3

The pathogenesis of neurodegenerative diseases caused by peripheral pathological proteins. After these peripheral pathological proteins enter the brain, they can activate microglia and astrocytes, triggering neuroinflammation, and are involved in the formation of Aβ plaques, neurofibrillary tangles, and Lewy bodies. They also lead to neuronal loss and degeneration. Additionally, disruption of the BBB by these proteins creates a vicious cycle, exacerbating neuropathology

PD

The gastrointestinal tract not only produces large amounts of Aβ and Tau but also generates α-syn, the pathological protein of PD [142, 143]. An increasing body of research has highlighted the importance of bidirectional communication along the gut-brain axis in the progression of PD [143]. Misfolded α-syn produced in the gut can propagate to the brain via the vagus nerve, where it can seed the formation of new Lewy bodies and contribute to the progression of the disease [144]. Furthermore, peripheral inflammation and gut dysbiosis may promote the misfolding and aggregation of α-syn, exacerbating the disease process [145].

The newly formed Lewy bodies lead to the degeneration of dopaminergic neurons in the substantia nigra, resulting in motor symptoms such as tremor, rigidity, and bradykinesia [146]. Instead of gut-derived α-syn, Yang et al. revealed that erythrocyte-derived α-syn can enter microglia and activate them, triggering neuroinflammation [139]. Additionally, erythrocytic α-syn can accumulate in astrocytic end-feet, regulating glutamate homeostasis and the release of vascular endothelial growth factor A (VEGFA) and inducible nitric oxide synthase (iNOS). Elevated levels of VEGFA and iNOS can disrupt the tight junctions between microvascular endothelial cells, compromising the BBB integrity [140, 147]. These effects collectively contribute to the occurrence and progression of neurodegenerative diseases such as PD (Fig. 3).

Other neurodegenerative diseases

The roles of peripheral Aβ, Tau, α-syn, and TDP-43 in other neurodegenerative diseases, such as ALS, FTD, Lewy body dementia, and MSA, are poorly understood. Furthermore, although TDP-43 is widely expressed in peripheral tissues and shows abnormal expression in the muscles of ALS patients [148], no studies have provided evidence that TDP-43 can enter the brain. Given the similarities in protein misfolding and aggregation across these disorders, it is plausible that peripheral sources of these proteins may also contribute to other neurodegenerative pathogenesis [149]. However, further research is needed to confirm this.

Peripheral immune dysfunction induced by peripheral proteinopathies in neurodegenerative diseases

In addition to causing neuropathology in the brain, recent studies have found that these peripheral pathological proteins can also induce dysfunction of the peripheral immune system, thereby exacerbating the progression of neurodegenerative diseases. Recent studies have shown that peripheral accumulation of pathological proteins can activate the peripheral immune system, causing chronic inflammation and accelerating the progression of neurodegenerative diseases [150]. Research has found presence of a large amount of Aβ aggregates and reduced levels of monocytes in the plasma even before the prodromal stage of AD [151], which may be related to an increased number and activity of monocyte-derived osteoclasts in the blood [152]. Though still controversial (see early discussion), peripheral Aβ has been shown to contribute to the pathogenesis of AD by activating innate immune cells, which promote the secretion of pro-inflammatory cytokines and molecules that increase BBB permeability [153]. Single-cell RNA-seq revealed that the abundance of human peripheral immune cells is regulated by Tau-related pathophysiological changes [154]. A study involving 1107 participants indicates that Aβ and Tau may be associated with changes in neutrophils and lymphocytes in the periphery and the neutrophil/lymphocyte ratio in AD [155], although the mechanism remains to be investigated. Our research team demonstrated that oligomeric α-syn derived from red blood cells can enter monocytes via receptor-mediated endocytosis, activating LRRK2 and leading to immune hyperactivation of monocytes in the circulatory system [156]. TDP-43 is also expressed in immune cells in peripheral blood, particularly in monocytes [157]. EVs containing TDP-43 induce activation of peripheral monocytes and functional deregulation of monocytes in ALS [158]. These studies underscore the important role of peripheral pathological proteins in neurodegenerative diseases not only by modulating central pathology but also through regulation of peripheral homeostasis, offering new approaches for the diagnosis and treatment of neurodegenerative disease.

Potential therapeutic targets

Given the potential roles of peripheral pathological proteins in the progression of neurodegenerative diseases, an important therapeutic strategy is to accelerate the clearance of these peripheral pathological proteins. Currently, several groups are attempting various approaches to reduce peripheral Aβ in order to improve AD pathology (Table 2). Monoclonal antibodies including Lecanemab and Donanemab have been verified to bind Aβ and enhance its clearance from the brain through several mechanisms, such as facilitating microglial phagocytosis, dissolving fibrillar Aβ, and activating the complement system [159,160,161,162]. However, these antibodies also bind to peripheral Aβ, facilitating Aβ efflux from CNS via a peripheral sink mechanism, without the need to cross the BBB to clear brain Aβ [163]. It is conceivable that after these antibodies neutralize Aβ in the blood, they may tilt the equilibrium of Aβ between the central and peripheral compartments, assisting the transport of Aβ from the brain to the peripheral blood via the LRP1 transporter [164, 165]. Sun et al. found that clearing Aβ from peripheral blood significantly reduces Aβ deposition in the brains of AD mice [166]. Rejuvenation of the peripheral immune system has also been confirmed to improve the pathology of AD [167]. It should be emphasized that approximately 60% of Aβ in the brain is cleared through its transport to peripheral tissues [33]. The liver and kidneys are the main organs that metabolize and clear Aβ and α-syn from blood. Several studies reported that accelerated clearance of peripheral pathological proteins such as Aβ inhibits disease progression. Conversely, insufficient metabolization in the liver and kidneys can exacerbate the pathology of AD [168, 169]. Moreover, the spleen is also involved in the physiological clearance of circulating Aβ in the periphery [170]. According to this clearance strategy, many techniques, including peritoneal dialysis and hemodialysis, have been explored for their efficacy in decreasing Aβ levels [171,172,173]. Additionally, some studies have discovered that the liver, as a major organ for clearing peripheral α-syn, could potentially improve the progression of PD pathology by enhancing the liver’s capacity to clear α-syn [92, 174].

Table 2 Summary of recent studies on clearing peripheral Aβ in AD

To effectively target peripheral proteinopathies, we must fully understand their pathological characteristics, transmission routes, and disease-causing mechanisms. This knowledge is essential for developing interventions that specifically target pathogenic forms while preserving the physiological functions of these proteins.

Another crucial strategy is to improve clearance of pathogenic proteins. Enhancing the functions of peripheral organs like the liver and kidneys, improving blood purification methods, and targeting lymphatic pathways are promising approaches to reducing brain proteinopathies. By mitigating the pathological effects of Aβ, Tau, and α-syn, these strategies could potentially slow disease progression.

Conclusions and perspectives

This review highlights the growing evidence for the peripheral origins of Aβ, Tau, α-syn, and TDP-43 and their entry into the CNS through various routes, including the bloodstream and neural pathways like the vagus nerve. Once in the brain, these proteins contribute to neuronal loss, synaptic inhibition, neuroinflammation, and BBB disruption, leading to the onset and progression of neurodegenerative diseases. Even without entering the brain, these proteinopathies can induce neuropathology by modulating the peripheral immune system. These findings challenge the traditional view of neurodegenerative diseases as solely CNS disorders and underscore the importance of considering the role of peripheral tissues and organs in the pathogenesis. Our review presents a new perspective on neurodegenerative diseases by considering them from the perspective of peripheral proteinopathies, offering new insights and strategies for the prevention and treatment of these diseases.

While our understanding of the involvement of peripheral pathological proteins in neurodegenerative diseases has advanced, many questions remain unanswered, including: (1) the mechanisms underlying the production and release of these proteins in both normal physiology and disease conditions, (2) the detailed pathways through which peripheral pathological proteins enter the brain, particularly the role of the vagus nerve, (3) the pathogenic mechanisms of these proteins in the brain, including their interactions with other pathological proteins, which is crucial for developing targeted therapies and preventive strategies against neurodegenerative diseases, and (4) the role of other peripheral amyloidogenic peptides, such as islet amyloid polypeptides (known as amylin) that can also spontaneously aggregate and cross the BBB, propagating into the brain and contributing to neuropathology [175,176,177].

Furthermore, efforts should focus on developing effective therapeutic strategies targeting peripheral pathological proteins, including drugs and interventions that specifically target these proteins, as well as strategies to modulate their production and release from peripheral tissues. Through these endeavors, we can deepen our understanding of the pathogenesis of neurodegenerative diseases and pave the way for novel approaches to the prevention and therapy of these debilitating disorders.

Availability of data and materials

Not applicable.

Abbreviations

CNS:

Central nervous system

AD:

Alzheimer’s disease

PD:

Parkinson’s disease

MSA:

Multiple system atrophy

ALS:

Amyotrophic lateral sclerosis

FTD:

Frontotemporal dementia

Aβ:

Amyloid-β

α-syn:

α-synuclein

TDP-43:

TAR DNA-binding protein 43

APP:

Amyloid precursor protein

BACE1:

β-Secretase

NCT:

Nicastrin

BBB:

Blood–brain barrier

EVs:

Extracellular vesicles

RBC-EVs:

Red blood cell-derived EVs

References

  1. Coukos R, Krainc D. Key genes and convergent pathogenic mechanisms in Parkinson disease. Nat Rev Neurosci. 2024;25(6):393–413.

    Article  CAS  PubMed  Google Scholar 

  2. Tetter S, Arseni D, Murzin AG, Buhidma Y, Peak-Chew SY, Garringer HJ, et al. TAF15 amyloid filaments in frontotemporal lobar degeneration. Nature. 2024;625(7994):345–51.

    Article  CAS  PubMed  Google Scholar 

  3. Humphrey J, Venkatesh S, Hasan R, Herb JT, de Paiva LK, Kucukali F, et al. Integrative transcriptomic analysis of the amyotrophic lateral sclerosis spinal cord implicates glial activation and suggests new risk genes. Nat Neurosci. 2023;26(1):150–62.

    Article  CAS  PubMed  Google Scholar 

  4. Dileep V, Boix CA, Mathys H, Marco A, Welch GM, Meharena HS, et al. Neuronal DNA double-strand breaks lead to genome structural variations and 3D genome disruption in neurodegeneration. Cell. 2023;186(20):4404–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Korczyn AD, Grinberg LT. Is Alzheimer disease a disease? Nat Rev Neurol. 2024;20(4):245–51.

    Article  PubMed  Google Scholar 

  6. Bashir S, Aiman A, Shahid M, Chaudhary AA, Sami N, Basir SF, et al. Amyloid-induced neurodegeneration: a comprehensive review through aggregomics perception of proteins in health and pathology. Ageing Res Rev. 2024;96: 102276.

    Article  PubMed  Google Scholar 

  7. Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell. 2023;30(5):512–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Esmaeili A, Eteghadi A, Landi FS, Yavari SF, Taghipour N. Recent approaches in regenerative medicine in the fight against neurodegenerative disease. Brain Res. 2024;1825: 148688.

    Article  CAS  PubMed  Google Scholar 

  9. Lau SF, Wu W, Wong HY, Ouyang L, Qiao Y, Xu J, et al. The VCAM1-ApoE pathway directs microglial chemotaxis and alleviates Alzheimer’s disease pathology. Nat Aging. 2023;3(10):1219–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zheng H, Shi C, Luo H, Fan L, Yang Z, Hu X, et al. Alpha-synuclein in parkinson’s disease: does a prion-like mechanism of propagation from periphery to the brain play a role? Neuroscientist. 2021;27(4):367–87.

    Article  CAS  PubMed  Google Scholar 

  11. Elia A, Parodi-Rullan R, Vazquez-Torres R, Carey A, Javadov S, Fossati S. Amyloid beta induces cardiac dysfunction and neuro-signaling impairment in the heart of an Alzheimer’s disease model. bioRxiv [Preprint]. 2023:2023.07.11.548558. doi: 10.1101/2023.07.11.548558.

  12. Balczon R, Lin MT, Lee JY, Abbasi A, Renema P, Voth SB, et al. Pneumonia initiates a tauopathy. FASEB J. 2021;35(9): e21807.

    Article  CAS  PubMed  Google Scholar 

  13. Ullah R, Park TJ, Huang X, Kim MO. Abnormal amyloid beta metabolism in systemic abnormalities and Alzheimer’s pathology: insights and therapeutic approaches from periphery. Ageing Res Rev. 2021;71: 101451.

    Article  CAS  PubMed  Google Scholar 

  14. Marquez A, Guernsey LS, Frizzi KE, Cundiff M, Constantino I, Muttalib N, et al. Tau associated peripheral and central neurodegeneration: identification of an early imaging marker for tauopathy. Neurobiol Dis. 2021;151: 105273.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gibbons C, Wang N, Rajan S, Kern D, Palma JA, Kaufmann H, et al. Cutaneous alpha-synuclein signatures in patients with multiple system atrophy and parkinson disease. Neurology. 2023;100(15):e1529–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Glenner GG, Wong CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 1984;120(3):885–90.

    Article  CAS  PubMed  Google Scholar 

  17. Masters CL, Multhaup G, Simms G, Pottgiesser J, Martins RN, Beyreuther K. Neuronal origin of a cerebral amyloid neurofibrillary tangles of Alzheimer’s disease contain the same protein as the amyloid of plaque cores and blood vessels. EMBO J. 1985;4(11):2757–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A. 1985;82(12):4245-9.

  19. Floden AM, Sohrabi M, Nookala S, Cao JJ, Combs CK. Salivary abeta secretion and altered oral microbiome in mouse models of AD. Curr Alzheimer Res. 2020;17(12):1133–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Roher AE, Esh CL, Kokjohn TA, Castano EM, Van Vickle GD, Kalback WM, et al. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 2009;5(1):18–29.

    Article  CAS  PubMed  Google Scholar 

  21. Yan P, Kim KW, Xiao Q, Ma X, Czerniewski LR, Liu H, et al. Peripheral monocyte-derived cells counter amyloid plaque pathogenesis in a mouse model of Alzheimer’s disease. J Clin Invest. 2022;132(11): e152565.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Tian Q, Bilgel M, Walker KA, Moghekar AR, Fishbein KW, Spencer RG, et al. Skeletal muscle mitochondrial function predicts cognitive impairment and is associated with biomarkers of Alzheimer’s disease and neurodegeneration. Alzheimers Dement. 2023;19(10):4436–45.

    Article  CAS  PubMed  Google Scholar 

  23. Torcinaro A, Ricci V, Strimpakos G, De Santa F, Middei S. Peripheral nerve impairment in a mouse model of Alzheimer’s disease. Brain Sci. 2021;11(9):1245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wu T, Chen L, Zhou L, Xu J, Guo K. Platelets transport beta-amyloid from the peripheral blood into the brain by destroying the blood-brain barrier to accelerate the process of Alzheimer’s disease in mouse models. Aging (Albany NY). 2021;13(5):7644–59.

    Article  CAS  PubMed  Google Scholar 

  25. Beura SK, Dhapola R, Panigrahi AR, Yadav P, Reddy DH, Singh SK. Redefining oxidative stress in Alzheimer’s disease: targeting platelet reactive oxygen species for novel therapeutic options. Life Sci. 2022;306:120855.

    Article  CAS  PubMed  Google Scholar 

  26. Bhattacharya A, Izzo A, Mollo N, Napolitano F, Limone A, Margheri F, et al. Inhibition of 37/67kDa Laminin-1 receptor restores APP maturation and reduces amyloid-beta in human skin fibroblasts from familial Alzheimer’s disease. J Pers Med. 2020;10(4):232.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Herrero-Labrador R, Trueba-Saiz A, Martinez-Rachadell L, Fernandez de Sevilla ME, Zegarra-Valdivia JA, Pignatelli J, et al. Circulating insulin-like growth factor I is involved in the effect of high fat diet on peripheral amyloid beta clearance. Int J Mol Sci. 2020;21(24):9675.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu BC, Tang TT, Lv LL. How tubular epithelial cell injury contributes to renal fibrosis. Adv Exp Med Biol. 2019;1165:233–52.

    Article  CAS  PubMed  Google Scholar 

  29. Jin J, Xu Z, Zhang L, Zhang C, Zhao X, Mao Y, et al. Gut-derived beta-amyloid: likely a centerpiece of the gut-brain axis contributing to Alzheimer’s pathogenesis. Gut Microbes. 2023;15(1):2167172.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Shi M, Chu F, Zhu F, Zhu J. Peripheral blood amyloid-β involved in the pathogenesis of Alzheimer’s disease via impacting on peripheral innate immune cells. J Neuroinflammation. 2024;21(1):5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bun S, Ito D, Tezuka T, Kubota M, Ueda R, Takahata K, et al. Performance of plasma Aβ42/40, measured using a fully automated immunoassay, across a broad patient population in identifying amyloid status. Alzheimers Res Ther. 2023;15(1):149.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yang J, Zhu B, Yin W, Han Z, Zheng C, Wang P, et al. Differentiating Aβ40 and Aβ42 in amyloid plaques with a small molecule fluorescence probe. Chem Sci. 2020;11(20):5238–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease - insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol. 2017;13(10):612–23.

    Article  CAS  PubMed  Google Scholar 

  34. Guerreiro RJ, Gustafson DR, Hardy J. The genetic architecture of Alzheimer's disease: beyond APP, PSENs and APOE. Neurobiol Aging. 2012;33(3):437–56.

    Article  Google Scholar 

  35. Liu N, Haziyihan A, Zhao W, Chen Y, Chao H. Trajectory of brain-derived amyloid beta in Alzheimer’s disease: where is it coming from and where is it going? Transl Neurodegener. 2024;13(1):42.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Joachim CL, Mori H, Selkoe DJ. Amyloid beta-protein deposition in tissues other than brain in Alzheimer’s disease. Nature. 1989;341(6239):226–30.

    Article  CAS  PubMed  Google Scholar 

  37. Koronyo Y, Salumbides BC, Black KL, Koronyo-Hamaoui M. Alzheimer’s disease in the retina: imaging retinal abeta plaques for early diagnosis and therapy assessment. Neurodegener Dis. 2012;10(1–4):285–93.

    Article  CAS  PubMed  Google Scholar 

  38. Troncone L, Luciani M, Coggins M, Wilker EH, Ho CY, Codispoti KE, et al. Abeta Amyloid pathology affects the hearts of patients with Alzheimer’s disease: mind the heart. J Am Coll Cardiol. 2016;68(22):2395–407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Romani M, Sorrentino V, Oh CM, Li H, de Lima TI, Zhang H, et al. NAD(+) boosting reduces age-associated amyloidosis and restores mitochondrial homeostasis in muscle. Cell Rep. 2021;34(3): 108660.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hur JY. Gamma-Secretase in Alzheimer’s disease. Exp Mol Med. 2022;54(4):433–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Guo X, Li H, Yan C, Lei J, Zhou R, Shi Y. Molecular mechanism of substrate recognition and cleavage by human gamma-secretase. Science. 2024;384(6700):1091–5.

    Article  CAS  PubMed  Google Scholar 

  42. Taylor HA, Przemylska L, Clavane EM, Meakin PJ. BACE1: More than just a β-secretase. Obes Rev. 2022;23(7): e13430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. De Strooper B, Karran E. New precision medicine avenues to the prevention of Alzheimer’s disease from insights into the structure and function of gamma-secretases. EMBO J. 2024;43(6):887–903.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Jurisch-Yaksi N, Sannerud R, Annaert W. A fast growing spectrum of biological functions of gamma-secretase in development and disease. Biochim Biophys Acta. 2013;1828(12):2815–27.

    Article  CAS  PubMed  Google Scholar 

  45. Pastore A, Raimondi F, Rajendran Temussi LPA. Why does the Aβ peptide of Alzheimer share structural similarity with antimicrobial peptides? Commun Biol. 2020;3(1):135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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(340):340ra72.

    Article  PubMed  Google Scholar 

  47. Tetz G, Tetz V. Bacterial extracellular DNA promotes β-amyloid aggregation. Microorganisms. 2021;9(6):1301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69(2):283–94.

    Article  CAS  PubMed  Google Scholar 

  49. Kowalski K, Mulak A. Brain-gut-microbiota axis in Alzheimer’s disease. J Neurogastroenterol Motil. 2019;25(1):48–60.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Xie J, Bruggeman A, De Nolf C, Vandendriessche C, Van Imschoot G, Van Wonterghem E, et al. Gut microbiota regulates blood-cerebrospinal fluid barrier function and Aβ pathology. Embo j. 2023;42(17): e111515.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen C, Liao J, Xia Y, Liu X, Jones R, Haran J, et al. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut. 2022;71(11):2233–52.

    Article  PubMed  Google Scholar 

  52. Tang Y, Zhang D, Robinson S, Zheng J. Inhibition of pancreatic cancer cells by different amyloid proteins reveals an inverse relationship between neurodegenerative diseases and cancer. Adv Biol (Weinh). 2023;7(8): e2300070.

    Article  PubMed  Google Scholar 

  53. Paris D, Townsend K, Quadros A, Humphrey J, Sun J, Brem S, et al. Inhibition of angiogenesis by Abeta peptides. Angiogenesis. 2004;7(1):75–85.

    Article  CAS  PubMed  Google Scholar 

  54. Li C, Liu K, Zhu J, Zhu F. The effects of high plasma levels of Aβ(1–42) on mononuclear macrophage in mouse models of Alzheimer’s disease. Immun Ageing. 2023;20(1):39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Taylor X, Clark IM, Fitzgerald GJ, Oluoch H, Hole JT, DeMattos RB, et al. Amyloid-β (Aβ) immunotherapy induced microhemorrhages are associated with activated perivascular macrophages and peripheral monocyte recruitment in Alzheimer’s disease mice. Mol Neurodegener. 2023;18(1):59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Poinsatte K, Smith EE, Torres VO, Ortega SB, Huebinger RM, Cullum CM, et al. T and B cell subsets differentially correlate with amyloid deposition and neurocognitive function in patients with amnestic mild cognitive impairment after one year of physical activity. Exerc Immunol Rev. 2019;25:34–49.

    PubMed  PubMed Central  Google Scholar 

  57. Yeapuri P, Machhi J, Lu Y, Abdelmoaty MM, Kadry R, Patel M, et al. Amyloid-β specific regulatory T cells attenuate Alzheimer’s disease pathobiology in APP/PS1 mice. Mol Neurodegener. 2023;18(1):97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci U S A. 1975;72(5):1858–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17(1):22–35.

    Article  CAS  Google Scholar 

  60. Goedert M, Spillantini MG, Crowther RA. Cloning of a big tau microtubule-associated protein characteristic of the peripheral nervous system. Proc Natl Acad Sci U S A. 1992;89(5):1983–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Boyne LJ, Tessler A, Murray M, Fischer I. Distribution of Big tau in the central nervous system of the adult and developing rat. J Comp Neurol. 1995;358(2):279–93.

    Article  CAS  PubMed  Google Scholar 

  62. Georgieff IS, Liem RK, Couchie D, Mavilia C, Nunez J, Shelanski ML. Expression of high molecular weight tau in the central and peripheral nervous systems. J Cell Sci. 1993;105(Pt 3):729–37.

    Article  CAS  PubMed  Google Scholar 

  63. Dugger BN, Harvey D, Beach TG, Adler CH. Peripheral tau as a biomarker for neurodegenerative diseases: is life on Earth, life on Mars? Brain. 2022;145(8):2629–31.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Chapelet G, Béguin N, Castellano B, Grit I, de Coppet P, Oullier T, et al. Tau expression and phosphorylation in enteroendocrine cells. Front Neurosci. 2023;17:1166848.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Solorzano A, Brady M, Bhatt N, Johnson A, Burgess B, Leyva H, et al. Central and peripheral tau retention modulated by an anti-tau antibody. bioRxiv [Preprint]. 2023:2023.08.17.553682. doi: 10.1101/2023.08.17.553682.

  66. Choi CS, Gwin M, Voth S, Kolb C, Zhou C, Nelson AR, et al. Cytotoxic tau released from lung microvascular endothelial cells upon infection with Pseudomonas aeruginosa promotes neuronal tauopathy. J Biol Chem. 2022;298(1): 101482.

    Article  CAS  PubMed  Google Scholar 

  67. Dugger BN, Whiteside CM, Maarouf CL, Walker DG, Beach TG, Sue LI, et al. The presence of select Tau species in human peripheral tissues and their relation to Alzheimer’s disease. J Alzheimers Dis. 2016;51(2):345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Alava B, Hery G, Sidhom S, Gutierrez-Monreal M, Prokop S, Esser KA, et al. Targeted brain-specific tauopathy compromises peripheral skeletal muscle integrity and function. Aging Brain. 2024;5:100110.

  69. Hart de Ruyter FJ, Morrema THJ, den Haan J, Netherlands Brain B, Twisk JWR, de Boer JF, et al. Phosphorylated tau in the retina correlates with tau pathology in the brain in Alzheimer’s disease and primary tauopathies. Acta Neuropathol. 2023;145(2):197–218.

    Article  CAS  PubMed  Google Scholar 

  70. Hamsafar Y, Chen Q, Borowsky AD, Beach TG, Serrano GE, Sue LI, et al. Biochemical analyses of tau and other neuronal markers in the submandibular gland and frontal cortex across stages of Alzheimer disease. Neurosci Lett. 2023;810: 137330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Betrie AH, Ayton S, Bush AI, Angus JA, Lei PCE, Wright. Evidence of a cardiovascular function for microtubule-associated protein Tau. J Alzheimers Dis. 2017;56(2):849–60.

    Article  CAS  PubMed  Google Scholar 

  72. Alquezar C, Arya S, Kao AW. Tau post-translational modifications: dynamic transformers of tau function, degradation, and aggregation. Front Neurol. 2021;11: 595532.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Dugger BN, Whiteside CM, Maarouf CL, Walker DG, Beach TG, Sue LI, et al. The presence of select tau species in human peripheral tissues and their relation to Alzheimer’s disease. J Alzheimers Dis. 2016;51(2):345–56.

    Article  CAS  Google Scholar 

  74. Ashton NJ, Brum WS, Di Molfetta G, Benedet AL, Arslan B, Jonaitis E, et al. Diagnostic accuracy of a plasma phosphorylated Tau 217 immunoassay for Alzheimer disease pathology. JAMA Neurol. 2024;81(3):255–63.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17(1):5–21.

    Article  PubMed  Google Scholar 

  76. Fischer I, Baas PW. Resurrecting the mysteries of big Tau. Trends Neurosci. 2020;43(7):493–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. DeC C, Revelli JP, Richman R, Yalamanchili HK, Kang MG, Han A, et al. The potentially protective role of an understudied tau isoform in Alzheimer’s disease. Alzheimers Dement. 2022;18: 059890.

    Google Scholar 

  78. Luciani M, Montalbano M, Troncone L, Bacchin C, Uchida K, Daniele G, et al. Big tau aggregation disrupts microtubule tyrosination and causes myocardial diastolic dysfunction: from discovery to therapy. Eur Heart J. 2023;44(17):1560–70.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Lim GB. Aggregation of big tau disrupts microtubules and causes diastolic dysfunction. Nat Rev Cardiol. 2023;20(7):441.

    Article  PubMed  Google Scholar 

  80. Renema P, Pittet JF, Brandon AP, Leal SM Jr, Gu S, Promer G, et al. Tau and Aβ42 in lavage fluid of pneumonia patients are associated with end-organ dysfunction: a prospective exploratory study. PLoS One. 2024;19(2): e0298816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Balczon R, Choi CS, deWeever A, Zhou C, Gwin MS, Kolb C, et al. Infection promotes Ser-214 phosphorylation important for generation of cytotoxic tau variants. FASEB J. 2023;37(7): e23042.

    Article  CAS  PubMed  Google Scholar 

  82. Alava B, Hery G, Sidhom S, Gutierrez-Monreal M, Prokop S, Esser KA, et al. Targeted brain-specific tauopathy compromises peripheral skeletal muscle integrity and function. Aging Brain. 2024;5: 100110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bridi JC, Hirth F. Mechanisms of α-synuclein induced synaptopathy in Parkinson’s disease. Front Neurosci. 2018;12:80.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Dutta S, Hornung S, Kruayatidee A, Maina KN, Del Rosario I, Paul KC, et al. α-Synuclein in blood exosomes immunoprecipitated using neuronal and oligodendroglial markers distinguishes Parkinson’s disease from multiple system atrophy. Acta Neuropathol. 2021;142(3):495–511.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Bridi JC, Hirth F. Mechanisms of alpha-Synuclein induced synaptopathy in Parkinson’s disease. Front Neurosci. 2018;12:80.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Norris EH, Giasson BI, Lee VM. Alpha-synuclein: normal function and role in neurodegenerative diseases. Curr Top Dev Biol. 2004;60:17–54.

    Article  CAS  PubMed  Google Scholar 

  87. Chahine LM, Beach TG, Brumm MC, Adler CH, Coffey CS, Mosovsky S, et al. In vivo distribution of α-synuclein in multiple tissues and biofluids in Parkinson disease. Neurology. 2020;95(9):e1267–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Veys L, Vandenabeele M, Ortuno-Lizaran I, Baekelandt V, Cuenca N, Moons L, et al. Retinal alpha-synuclein deposits in Parkinson’s disease patients and animal models. Acta Neuropathol. 2019;137(3):379–95.

    Article  CAS  PubMed  Google Scholar 

  89. Casini A, Mancinelli R, Mammola CL, Pannarale L, Chirletti P, Onori P, et al. Distribution of α-synuclein in normal human jejunum and its relations with the chemosensory and neuroendocrine system. Eur J Histochem. 2021;65(4):3310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Jiang W, Cheng Y, Wang Y, Wu J, Rong Z, Sun L, et al. Involvement of abnormal p-α-syn accumulation and TLR2-mediated inflammation of schwann cells in enteric autonomic nerve dysfunction of parkinson’s disease: an animal model study. Mol Neurobiol. 2023;60(8):4738–52.

    Article  CAS  PubMed  Google Scholar 

  91. Ma LY, Liu GL, Wang DX, Zhang MM, Kou WY, Feng T. Alpha-synuclein in peripheral tissues in parkinson’s disease. ACS Chem Neurosci. 2019;10(2):812–23.

    Article  CAS  PubMed  Google Scholar 

  92. Reyes JF, Ekmark-Léwen S, Perdiki M, Klingstedt T, Hoffmann A, Wiechec E, et al. Accumulation of alpha-synuclein within the liver, potential role in the clearance of brain pathology associated with Parkinson’s disease. Acta Neuropathol Commun. 2021;9(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep L, et al. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5(2):55–9.

    Article  CAS  PubMed  Google Scholar 

  94. Bartels T, Choi JG, Selkoe DJ. alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature. 2011;477(7362):107–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Beyer K. Mechanistic aspects of Parkinson’s disease: alpha-synuclein and the biomembrane. Cell Biochem Biophys. 2007;47(2):285–99.

    Article  CAS  PubMed  Google Scholar 

  96. Oliveira JT, Dakic V, Vitória G, Pedrosa C, Mendes M, Aragão L, et al. Oligomeric α-Synuclein induces skin degeneration in reconstructed human epidermis. Neurobiol Aging. 2022;113:108–17.

    Article  CAS  PubMed  Google Scholar 

  97. Han Y, Wu D, Wang Y, Xie J, Zhang Z. Skin alpha-synuclein deposit patterns: a predictor of Parkinson’s disease subtypes. EBioMedicine. 2022;80: 104076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Schmidhuber S, Scheiblhofer S, Weiss R, Cserepes M, Tóvári J, Gadermaier G, et al. A Novel C-type lectin receptor-targeted α-synuclein-based parkinson vaccine induces potent immune responses and therapeutic efficacy in mice. Vaccines (Basel). 2022;10(9):1432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of α-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci. 2013;14(1):38–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Fasano A, Visanji NP, Liu LW, Lang AE, Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2015;14(6):625–39.

    Article  CAS  PubMed  Google Scholar 

  101. Yang Y, Stewart T, Zhang C, Wang P, Xu Z, Jin J, et al. Erythrocytic α-synuclein and the gut microbiome: kindling of the gut-brain axis in Parkinson’s disease. Mov Disord. 2024;39(1):40–52.

    Article  CAS  PubMed  Google Scholar 

  102. Challis C, Hori A, Sampson TR, Yoo BB, Challis RC, Hamilton AM, et al. Gut-seeded alpha-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat Neurosci. 2020;23(3):327–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Tziortzouda P, VanDen Bosch L, Hirth F. Triad of TDP43 control in neurodegeneration: autoregulation, localization and aggregation. Nat Rev Neurosci. 2021;22(4):197–208.

    Article  CAS  PubMed  Google Scholar 

  104. de Boer EMJ, Orie VK, Williams T, Baker MR, De Oliveira HM, Polvikoski T, et al. TDP-43 proteinopathies: a new wave of neurodegenerative diseases. J Neurol Neurosurg Psychiatry. 2020;92(1):86–95.

    Article  PubMed  Google Scholar 

  105. Wang W, Wang L, Lu J, Siedlak SL, Fujioka H, Liang J, et al. The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med. 2016;22(8):869–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Nishino K, Watanabe S, Shijie J, Murata Y, Oiwa K, Komine O, et al. Mice deficient in the C-terminal domain of TAR DNA-binding protein 43 develop age-dependent motor dysfunction associated with impaired Notch1-Akt signaling pathway. Acta Neuropathol Commun. 2019;7(1):118.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Lopez-Carbonero JI, Garcia-Toledo I, Fernandez-Hernandez L, Bascunana P, Gil-Moreno MJ, Matias-Guiu JA, et al. In vivo diagnosis of TDP-43 proteinopathies: in search of biomarkers of clinical use. Transl Neurodegener. 2024;13(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  108. D’Agostino C, Nogalska A, Engel WK, Askanas V. In sporadic inclusion body myositis muscle fibres TDP-43-positive inclusions are less frequent and robust than p62 inclusions, and are not associated with paired helical filaments. Neuropathol Appl Neurobiol. 2011;37(3):315–20.

    Article  PubMed  Google Scholar 

  109. Weihl CC, Temiz P, Miller SE, Watts G, Smith C, Forman M, et al. TDP-43 accumulation in inclusion body myopathy muscle suggests a common pathogenic mechanism with frontotemporal dementia. J Neurol Neurosurg Psychiatry. 2008;79(10):1186–9.

    Article  CAS  PubMed  Google Scholar 

  110. Zhang QJ, Lin J, Wang YL, Chen L, Ding Y, Zheng FZ, et al. Detection of pTDP-43 via routine muscle biopsy: a promising diagnostic biomarker for amyotrophic lateral sclerosis. Brain Pathol. 2024;34(6):e13261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Mori F, Tada M, Kon T, Miki Y, Tanji K, Kurotaki H, et al. Phosphorylated TDP-43 aggregates in skeletal and cardiac muscle are a marker of myogenic degeneration in amyotrophic lateral sclerosis and various conditions. Acta Neuropathol Commun. 2019;7(1):165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Cykowski MD, Powell SZ, Appel JW, Arumanayagam AS, Rivera AL, Appel SH. Phosphorylated TDP-43 (pTDP-43) aggregates in the axial skeletal muscle of patients with sporadic and familial amyotrophic lateral sclerosis. Acta Neuropathol Commun. 2018;6(1):28.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Kurashige T, Morino H, Murao T, Izumi Y, Sugiura T, Kuraoka K, et al. TDP-43 accumulation within intramuscular nerve bundles of patients with amyotrophic lateral sclerosis. JAMA Neurol. 2022;79(7):693–701.

    Article  PubMed  Google Scholar 

  114. Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11(3):155–9.

    Article  CAS  PubMed  Google Scholar 

  115. Jaunmuktane Z, Brandner S. Invited review: the role of prion-like mechanisms in neurodegenerative diseases. Neuropathol Appl Neurobiol. 2020;46(6):522–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Prusiner SB. Biology and genetics of prions causing neurodegeneration. Annu Rev Genet. 2013;47:601–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Gomez-Gutierrez R, Morales R. The prion-like phenomenon in Alzheimer’s disease: evidence of pathology transmission in humans. PLoS Pathog. 2020;16(10): e1009004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 2013;501(7465):45–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wilson DM 3rd, Cookson MR, Van Den Bosch L, Zetterberg H, Holtzman DMDewachter I. Hallmarks of neurodegenerative diseases. Cell. 2023;186(4):693–714.

    Article  CAS  PubMed  Google Scholar 

  120. 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(19):4209–23.

    Article  CAS  PubMed  Google Scholar 

  121. Ahn EH, Kang SS, Liu X, Chen G, Zhang Z, Chandrasekharan B, et al. Initiation of Parkinson’s disease from gut to brain by delta-secretase. Cell Res. 2020;30(1):70–87.

    Article  PubMed  Google Scholar 

  122. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, et al. Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models parkinson’s disease. Neuron. 2019;103(4):627–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Lai PH, Wang TH, Zhang NY, Wu KC, Yao CJ, Lin CJ. Changes of blood-brain-barrier function and transfer of amyloid beta in rats with collagen-induced arthritis. J Neuroinflammation. 2021;18(1):35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sheng L, Stewart T, Yang D, Thorland E, Soltys D, Aro P, et al. Erythrocytic alpha-synuclein contained in microvesicles regulates astrocytic glutamate homeostasis: a new perspective on Parkinson’s disease pathogenesis. Acta Neuropathol Commun. 2020;8(1):102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Sun Y, Sommerville NR, Liu JYH, Ngan MP, Poon D, Ponomarev ED, et al. Intra-gastrointestinal amyloid-beta1-42 oligomers perturb enteric function and induce Alzheimer’s disease pathology. J Physiol. 2020;598(19):4209–23.

    Article  CAS  PubMed  Google Scholar 

  126. Neuhuber WL, Berthoud H-R. Functional anatomy of the vagus system–emphasis on the somato-visceral interface. Auton Neurosci. 2021;236: 102887.

    Article  PubMed  PubMed Central  Google Scholar 

  127. 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(17): e106320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Uemura N, Yagi H, Uemura MT, Hatanaka Y, Yamakado H, Takahashi R. Inoculation of α-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol Neurodegener. 2018;13(1):21.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Yang Y, Stewart T, Zhang C, Wang P, Xu Z, Jin J, et al. Erythrocytic alpha-synuclein and the gut microbiome: kindling of the gut-brain axis in Parkinson’s disease. Mov Disord. 2024;39(1):40–52.

    Article  CAS  PubMed  Google Scholar 

  130. 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(1):2167172.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Xiang J, Tang J, Kang F, Ye J, Cui Y, Zhang Z, et al. Gut-induced alpha-synuclein and Tau propagation initiate Parkinson’s and Alzheimer’s disease co-pathology and behavior impairments. Neuron. 2024;112(21):3585.

    Article  CAS  PubMed  Google Scholar 

  132. Arotcarena ML, Dovero S, Prigent A, Bourdenx M, Camus S, Porras G, et al. Bidirectional gut-to-brain and brain-to-gut propagation of synucleinopathy in non-human primates. Brain. 2020;143(5):1462–75.

    Article  PubMed  Google Scholar 

  133. Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS, Chui HC, et al. Vascular dysfunction—the disregarded partner of Alzheimer’s disease. Alzheimers Dement. 2019;15(1):158–67.

    Article  Google Scholar 

  134. Yamazaki Y, Kanekiyo T. Blood-brain barrier dysfunction and the pathogenesis of Alzheimer’s disease. Int J Mol Sci. 2017;18(9):1965.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Akhter F, Akhter A, Kesari KK, Javed R, Ruokolainen J, Vuorinen T. RAGE exacerbate amyloid beta (Aβ) induced alzheimer pathology: A systemic overview. . In: Kesari, K. (eds) Networking of Mutagens in Environmental Toxicology. Environmental Science and Engineering. Springer, Cham; 2019: 159–170. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-3-319-96511-6_9

  136. Liu Q, Li D, Pan X, Liang Y. Targeted therapy using engineered extracellular vesicles: principles and strategies for membrane modification. J Nanobiotechnology. 2023;21(1):334.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Ha JY, Seok J, Kim SJ, Jung HJ, Ryu KY, Nakamura M, et al. Periodontitis promotes bacterial extracellular vesicle-induced neuroinflammation in the brain and trigeminal ganglion. PLoS Pathog. 2023;19(10): e1011743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Makrygianni EA, Chrousos GP. Extracellular vesicles and the stress system. Neuroendocrinology. 2023;113(2):120–67.

    Article  CAS  PubMed  Google Scholar 

  139. Matsumoto J, Stewart T, Sheng L, Li N, Bullock K, Song N, et al. Transmission of alpha-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: another mechanism for initiation and progression of Parkinson’s disease? Acta Neuropathol Commun. 2017;5(1):71.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Sheng L, Stewart T, Yang D, Thorland E, Soltys D, Aro P, et al. Erythrocytic α-synuclein contained in microvesicles regulates astrocytic glutamate homeostasis: a new perspective on Parkinson’s disease pathogenesis. Acta Neuropathol Commun. 2020;8(1):102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Bruno M, Bonomi CG, Ricci F, Di Donna MG, Mercuri NB, Koch G, et al. Blood–brain barrier permeability is associated with different neuroinflammatory profiles in Alzheimer’s disease. Eur J Neurol. 2024;31(1): e16095.

    Article  PubMed  Google Scholar 

  142. Kim Y, McInnes J, Kim J, Liang YHW, Veeraragavan S, Garza AR, et al. Olfactory deficit and gastrointestinal dysfunction precede motor abnormalities in alpha-Synuclein G51D knock-in mice. Proc Natl Acad Sci U S A. 2024;121(39): e2406479121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Musgrove RE, Helwig M, Bae E-J, Aboutalebi H, Lee S-J, Ulusoy A, et al. Oxidative stress in vagal neurons promotes parkinsonian pathology and intercellular α-synuclein transfer. J Clin Investig. 2019;129(9):3738–53.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Kim S, Kwon S-H, Kam T-I, Panicker N, Karuppagounder SS, Lee S, et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron. 2019;103(4):627–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Lei Q, Wu T, Wu J, Hu X, Guan Y, Wang Y, et al. Roles of α-synuclein in gastrointestinal microbiome dysbiosis-related Parkinson’s disease progression. Mol Med Rep. 2021;24(4):1–14.

    Article  Google Scholar 

  146. Stefanis L. α-Synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2(2): a009399.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Lan G, Wang P, Chan RB, Liu Z, Yu Z, Liu X, et al. Astrocytic VEGFA: an essential mediator in blood-brain-barrier disruption in Parkinson’s disease. Glia. 2022;70(2):337–53.

    Article  CAS  PubMed  Google Scholar 

  148. Zhang QJ, Lin J, Wang YL, Chen L, Ding Y, Zheng FZ, et al. Detection of pTDP-43 via routine muscle biopsy: a promising diagnostic biomarker for amyotrophic lateral sclerosis. Brain Pathol. 2024;34(6):13261.

    Article  Google Scholar 

  149. Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018;21(10):1332–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Mary A, Mancuso R, Heneka MT. Immune activation in Alzheimer disease. Annu Rev Immunol. 2024;42(1):585–613.

    Article  CAS  PubMed  Google Scholar 

  151. Juul-Madsen K, Parbo P, Ismail R, Ovesen PL, Schmidt V, Madsen LS, et al. Amyloid-beta aggregates activate peripheral monocytes in mild cognitive impairment. Nat Commun. 2024;15(1):1224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Wu B, Chen M, Meng L, Tian Q, Dong Z. Osteoclasts link dysregulated peripheral degradation processes and accelerated progression in Alzheimer’s disease. J Alzheimers Dis. 2024;99(2):773–85.

    Article  CAS  PubMed  Google Scholar 

  153. Shi M, Chu F, Zhu F, Zhu J. Peripheral blood amyloid-beta involved in the pathogenesis of Alzheimer’s disease via impacting on peripheral innate immune cells. J Neuroinflammation. 2024;21(1):5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sirkis DW, Warly Solsberg C, Johnson TP, Bonham LW, Sturm VE, Lee SE, et al. Single-cell RNA-seq reveals alterations in peripheral CX3CR1 and nonclassical monocytes in familial tauopathy. Genome Med. 2023;15(1):53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Hou JH, Ou YN, Xu W, Zhang PF, Tan L, Yu JT, et al. Association of peripheral immunity with cognition, neuroimaging, and Alzheimer’s pathology. Alzheimers Res Ther. 2022;14(1):29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Liu Z, Chan RB, Cai Z, Liu X, Wu Y, Yu Z, et al. Alpha-Synuclein-containing erythrocytic extracellular vesicles: essential contributors to hyperactivation of monocytes in Parkinson’s disease. J Neuroinflammation. 2022;19(1):53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Quek H, Cuni-Lopez C, Stewart R, Colletti T, Notaro A, Nguyen TH, et al. ALS monocyte-derived microglia-like cells reveal cytoplasmic TDP-43 accumulation, DNA damage, and cell-specific impairment of phagocytosis associated with disease progression. J Neuroinflammation. 2022;19(1):58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zondler L, Feiler MS, Freischmidt A, Ruf WP, Ludolph AC, Danzer KM, et al. Impaired activation of ALS monocytes by exosomes. Immunol Cell Biol. 2017;95(2):207–14.

    Article  CAS  PubMed  Google Scholar 

  159. Swanson CJ, Zhang Y, Dhadda S, Wang J, Kaplow J, Lai RYK, et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer’s disease with lecanemab, an anti-Aβ protofibril antibody. Alzheimers Res Ther. 2021;13(1):80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, et al. Lecanemab in early alzheimer’s disease. N Engl J Med. 2023;388(1):9–21.

    Article  PubMed  Google Scholar 

  161. Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, et al. Donanemab in early symptomatic alzheimer disease: the trailblazer-ALZ 2 randomized clinical trial. JAMA. 2023;330(6):512–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Pontecorvo MJ, Lu M, Burnham SC, Schade AE, Dage JL, Shcherbinin S, et al. Association of donanemab treatment with exploratory plasma biomarkers in early symptomatic alzheimer disease: a secondary analysis of the trailblazer-ALZ randomized clinical trial. JAMA Neurol. 2022;79(12):1250–9.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Loeffler DA. Antibody-mediated clearance of brain amyloid-β: mechanisms of action, effects of natural and monoclonal anti-Aβ antibodies, and downstream effects. J Alzheimers Dis Rep. 2023;7(1):873–99.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. Clearance of alzheimer’s amyloid-ss(1–40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106(12):1489–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Deane R, Wu Z, Sagare A, Davis J, Du Yan S, Hamm K, et al. LRP/amyloid beta-peptide interaction mediates differential brain efflux of Abeta isoforms. Neuron. 2004;43(3):333–44.

    Article  CAS  PubMed  Google Scholar 

  166. 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(10):5568–77.

    Article  CAS  PubMed  Google Scholar 

  167. Sun PY, Liu J, Hu JN, Tu YF, Jiang Q, Jia YJ, et al. Rejuvenation of peripheral immune cells attenuates Alzheimer’s disease-like pathologies and behavioral deficits in a mouse model. Sci Adv. 2024;10(22):1123.

    Article  Google Scholar 

  168. 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(6):717–31.

    Article  CAS  PubMed  Google Scholar 

  169. 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(10):6074–82.

    Article  CAS  PubMed  Google Scholar 

  170. Yu ZY, Chen DW, Tan CR, Zeng GH, He CY, Wang J, et al. Physiological clearance of Aβ by spleen and splenectomy aggravates Alzheimer-type pathogenesis. Aging Cell. 2022;21(1): e13533.

    Article  CAS  PubMed  Google Scholar 

  171. Kitaguchi N, Kato T, Matsunaga S, Hirano K, Iwata K, Kawaguchi K, et al. Removal of blood amyloid-β with hemodialysis reduced brain amyloid-β, confirmed by brain imaging: a case report. Neuropsychiatr Dis Treat. 2018;14:2931–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Jin WS, Shen LL, Bu XL, Zhang WW, Chen SH, Huang ZL, 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):207–20.

    Article  CAS  PubMed  Google Scholar 

  173. Urayama A, Moreno-Gonzalez I, Morales-Scheihing D, Kharat V, Pritzkow S, Soto C. Preventive and therapeutic reduction of amyloid deposition and behavioral impairments in a model of Alzheimer’s disease by whole blood exchange. Mol Psychiatry. 2022;27(10):4285–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Zhu C, Zhu J, Xiang Y, Bu XL, JinWang WSYJ. A conceptual study on the peripheral clearance of brain-derived α-synuclein in humans. J Alzheimers Dis. 2022;90(4):1485–92.

    Article  CAS  PubMed  Google Scholar 

  175. Mukherjee A, Morales-Scheihing D, Butler PC, Soto C. Type 2 diabetes as a protein misfolding disease. Trends Mol Med. 2015;21(7):439–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Meng L, Li Y, Liu C, Zhang G, Chen J, Xiong M, et al. Islet amyloid polypeptide triggers alpha-synuclein pathology in Parkinson’s disease. Prog Neurobiol. 2023;226: 102462.

    Article  CAS  PubMed  Google Scholar 

  177. Bortoletto AS, Parchem RJ. A pancreatic player in dementia: pathological role for islet amyloid polypeptide accumulation in the brain. Neural Regen Res. 2023;18(10):2141–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

All figures are modified from BioRender (https://www.biorender.com/).

Funding

This work was supported by National Natural Science Foundation of China under Grant 82020108012 and 82371250, Natural Science Foundation of Zhejiang Province under Grant LY24H090006 and LZ23H090002, Key R&D Program of Zhejiang under Grant 2024C03098 and 2024SSYS0018, and Natural Science Foundation of Shandong Province under Grant ZR2022QH177.

Author information

Authors and Affiliations

Authors

Contributions

BX and JZ conceptualized and designed the review. BX and XL reviewed the pieces of literature and drafted the manuscript. XL prepared the figures. BX, XL, YY, JYY, JC, ZX, KQY, and JZ revised the final version of the manuscript. All authors contributed to the discussion and approved the final manuscript.

Corresponding author

Correspondence to Jing Zhang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, B., Lei, X., Yang, Y. et al. Peripheral proteinopathy in neurodegenerative diseases. Transl Neurodegener 14, 2 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-024-00461-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-024-00461-6

Keywords