Skip to main content

Pathological mechanisms of neuroimmune response and multitarget disease-modifying therapies of mesenchymal stem cells in Parkinson’s disease

Abstract

Parkinson's disease (PD) is a neurodegenerative disease characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN); the etiology and pathological mechanism of the disease are still unclear. Recent studies have shown that the activation of a neuroimmune response plays a key role in the development of PD. Alpha-synuclein (α-Syn), the primary pathological marker of PD, can gather in the SN and trigger a neuroinflammatory response by activating microglia which can further activate the dopaminergic neuron’s neuroimmune response mediated by reactive T cells through antigen presentation. It has been shown that adaptive immunity and antigen presentation processes are involved in the process of PD and further research on the neuroimmune response mechanism may open new methods for its prevention and therapy. While current therapeutic regimens are still focused on controlling clinical symptoms, applications such as immunoregulatory strategies can delay the symptoms and the process of neurodegeneration. In this review, we summarized the progression of the neuroimmune response in PD based on recent studies and focused on the use of mesenchymal stem cell (MSC) therapy and challenges as a strategy of disease-modifying therapy with multiple targets.

Introduction

Parkinson's disease (PD) is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra [1, 2]. The main clinical manifestations are quiescent tremors, myotonia, tardiness, and abnormal postural gait [3, 4]. Abnormalities of the immune system are also considered an essential component of PD susceptibility and progression, and this area of research has received increasing attention over the past decade. PD is characterized by the death of dopaminergic neurons containing Lewy bodies (LB) composed mainly of alpha-synuclein (α-Syn) in the substantia nigra (SN), which provide the final diagnostic and pathological features for postmortem examinations [5, 6]. The α-Syn protein is a small acidic synaptic protein made up of 140 amino acids with a tendency to misfold and aggregate [7]. During the course of the disease, the major constituent of LB is misfolded α-Syn, which spreads to different brain regions in a prion-like fashion [8]. Microglia are the first immune defense system of the human brain and one of the main cell types involved in the inflammatory response of the central nervous system [9, 10]. The misfolded α-Syn protein can bind to and activate microglial cells through receptors on the surface of microglial cells, leading to a series of persistent inflammatory responses [11, 12].

Due to the lack of understanding of the cause of PD, symptomatic clinical therapy has traditionally focused on using medicines like levodopa and brain surgery. Besides the adverse effects associated with using long-term oral levodopa, drug resistance is the critical reason that renders the treatment ineffective. While they ameliorate patients' symptoms to some extent, surgeries like neuronuclear destruction and deep brain stimulation can cause irreversible neurological damage [13, 14]. In recent years, some vaccines and antibody formulations targeting α-Syn by active and passive immunization have been developed according to the main pathological markers of PD, and preliminary efficacy and safety assessments have been achieved [15,16,17]. Nerve repair, replacement, and regeneration have become possible with the development of stem cell technology and progress has been made in treating PD with stem cell transplantation. Mesenchymal stem cells (MSCs) are adult stem cells with paracrine, immune regulation, and multidirectional differentiation potential [18,19,20]. It was previously believed that the main pathogenic mechanism of PD was the loss of dopaminergic neurons and most researchers considered that the therapeutic effect of MSCs on PD was attributable to their cell replacement ability [21, 22]. The research focus of MSCs has shifted from cell replacement to multitarget therapy such as paracrine and immune regulation [23, 24]. The results of the studies also show that MSCs can accelerate the clearance and degradation of α-Syn through multitarget-modifying, such as modulating microglia activation, autophagic pathway, endocytosis, and protease secretion [25,26,27,28]. Research in the emerging field of PD immunomodulation provides an opportunity to identify new therapeutic targets and strategies to attenuate or reverse neurodegenerative changes. This review provides a concise overview of the development of neuroimmune responses and immune regulations in PD vaccine and antibody preparations targeting α-Syn through active and passive immunization, and the latest theoretical research and efficacy evaluation results of MSCs for the treatment of PD through multitarget disease modifications.

The mechanism of the neuroimmune response in PD

The α-Syn prion-like spreading

The aggregation of misfolded proteins in the central nervous system is a crucial hallmark of several age-related neurodegenerative diseases, including PD, Alzheimer's disease, and amyotrophic lateral sclerosis [29]. These diseases share key biochemical and biophysical characteristics with prion diseases. PD patients are believed to have a neuropathological basis denoted by the presence of LBs, which mostly comprise α-Syn inclusions. Pathological aggregation of α-Syn and its propagation through synaptic coupling are increasingly recognized as the basis for the pathophysiological progression of PD and related synuclein diseases [29]. Although the precise molecular mechanisms responsible for pathological accumulation and diffusion of α-Syn in the central nervous system are unclear, there is increasing evidence that misfolding and/or neuronal internalization of α-Syn promotes conformational template formation of endogenous α-Syn in monocytes by mechanisms similar to prion formation [30]. Microglia are macrophage-like populations of the central nervous system, which remain quiescent until injury or infection activates the cells to perform inflammatory functions and antigen-presenting cell (APC) functions [31]. The α-Syn aggregates in the central nervous system and can trigger a neuroinflammatory response by activating microglia, which can further activate the dopaminergic neuron neuroimmune response mediated by reactive T cells through antigen presentation [4, 31, 32].

Innate and adaptive immunity

There are innate and adaptive parts of the immune system that work together to fight infection; however, when the immune system is maladjusted, the immune response can be a significant trigger of the disease. The innate immune system is the body’s first line of defense against the invasion of pathogens and consists of various cells that perform functions such as phagocytosis and antigen presentation. Due to the blood–brain barrier (BBB), the human brain has long been considered an immune-privileged organ. Prior research on the immune response of neurodegenerative diseases mainly focused on the innate immune system including dendritic cells, macrophages, and microglia, which can be activated by recognizing pathogen-associated molecular patterns and pattern-recognition receptors of endogenous damage-associated molecular patterns (such as α-Syn) [33,34,35]. The adaptive immune system of B and T lymphocytes produces a highly specific, targeted response to a wide variety of intracellular and extracellular infections [36]. CD4 + or CD8 + T cells are naive until their T cell receptors (TCRs) recognize the specific antigen presented by antigen-presenting cells via major histocompatibility complex (MHC) molecules [37]. In general, endogenous antigens are presented to CD8 + T cells by MHC-I, while exogenous resistance components are presented to CD4 + T cells by MHC-II [38]. All cells in the human body express MHC-I and can activate CD8 + T cells, while only specialized antigen-presenting cells including dendritic cells, macrophages, and microglia express MHC-II and have the ability to activate CD4 + T cells [38]. Once activated in secondary lymphoid organs, naive T cells differentiate into effector cells with specific functions to adapt to infection. CD8 + T cells can differentiate into cytotoxic T lymphocytes and induce apoptosis of infected cells without affecting adjacent healthy cells [39]. CD4 + T cells differentiate into helper T cell subtypes (Th1, Th2, and Th17 cells), which can produce various cytokines that help B lymphocytes [39]. Once activated by their specific antigen, primitive B lymphocytes can differentiate into plasma cells with the help of T helper cells, which produce antibodies specific to the antigen and promote clearance through phagocytosis [39].

The recent discovery of meningeal lymphatic vessels has directly overturned the traditional idea of brain immune privilege [40, 41]. There is evidence that central nervous system antigens can interact with lymphocytes and antigen-presenting cells through meningeal lymphatic pathways, which suggests that both innate and adaptive immune responses may be involved in the development of neurodegenerative disease [40,41,42]. Many studies have shown that persistent inflammatory response, microglial cell activation, and T cell infiltration are common characteristics of PD models and patients, and these factors play a vital role in the degeneration of dopaminergic neurons [43,44,45]. Seeking new therapeutic strategies to suppress neuroinflammatory responses can prevent or delay the loss of neurons in the SN and prevent disease progression. We have delineated an overview of the development of the neuroimmune response to PD as provided below.

Mechanisms and roles of α-Syn in microglia cells

PD and brain–gut–microbe axis

The gut flora and other microbes control integral functions of immune cells in the gut, periphery, and brain. The presence of intestinal inflammation or gastrointestinal abnormalities such as constipation and diarrhea often precedes movement disorders in PD patients for many years [31, 46]. According to Braak’s hypothesis, abnormal α-Syn accumulation begins in the gut and spreads to the brain through the vagus nerve. It appears early in the enteric nervous system, glossopharyngeal nerve, and vagus nerve. It is possible that shaving the vagus nerve has a reduced risk of PD (Fig. 1) [47]. The α-Syn travels in the brain in a prion-like fashion between different neurons and brain regions. Although there is no doubt that the gastrointestinal tract is involved in the occurrence and development of PD, there are still limitations in the study of PD and the brain-gut-microbe axis. Biopsy of the SN does not explain the timing of α-Syn deposition, systemic autopsy could not explain the dynamic process of α-Syn deposition, and animal experiments do not perfectly simulate the occurrence and development of PD. Even the mechanism of misfolding and deposition of α-Syn is not clear in vivo, so it is too early to directly implicate misfolding of α-Syn in the gastrointestinal tract as the origin of PD. Studies have found that α-Syn is expressed presynaptically in the perinuclear area in the central nervous system, and there is a dynamic balance between production and degradation in healthy people [48]. The ubiquitin proteasome system (UPS) and autophagy–lysosome pathway (ALP) are two critical intracellular mechanisms for removing misfolded or aged proteins, so they have a significant impact on maintaining α-Syn levels. The UPS selectively degrades intracellular, membrane, misfolded, and damaged proteins [49,50,51]. Parkin and UCHL1, with two genetic mutations found in PD, affect the function of the UPS, so α-Syn does not degrade effectively and forms Lewy bodies, and the ALP pathway is a compensatory degradation mode for the damaged UPS [52, 53]. Neuronal cells have only a limited ability to remove α-Syn, and misfolding and/or neuronal internalization of α-Syn promote conformational template formation of endogenous α-Syn monocytes by mechanisms similar to prion-like spreading [30].

Fig. 1
figure 1

Mechanisms and roles of microglia activated by α-Syn on T cells in PD. Pathologic factors cause abnormal α-Syn accumulation in the brain parenchyma (1). Toll-like receptors on microglia can recognize misfolded α-Syn and promote the transformation of non-activated microglia into activated microglia and antigen-presenting cells (2). Proinflammatory cytokines and ROS secreted by microglia amplify neuroinflammatory responses in the brain and hasten the death of dopaminergic neurons (3). α-Syn aggregation in microglia activates NLRP3 inflammasomes, amplifying the inflammatory response and damaging dopaminergic neurons (4). The blood–brain barrier is compromised with age and disease, allowing peripheral blood immune components such as T cells to infiltrate the brain parenchyma (5). Activated microglia increase the secretion of inflammatory chemokines like RANTES and eotaxin, increasing CD8 + T cell infiltration and the cytotoxic response to dopaminergic neurons (6, 7). T lymphocyte-induced dopaminergic neuron death was mediated by the IL-17-IL-17R signaling pathway, which then activated the downstream NF-B signaling pathway (8). The images in this figure were downloaded from ProteinLounge.com with permission to use them

PD and microglial activation

Microglia, as the first line of defense of the immune system, similar to macrophages in the central nervous system, can respond quickly to invading pathogens, changes in the physiological microenvironment, and damage to the central nervous system, playing a centric role in the central inflammatory response [10, 54, 55]. Acute inflammatory reactions mediated by microglia are generally thought to benefit neuronal survival via the predominant phenotype M2. Nevertheless, the long-term and excessive activation of microglia can result in chronic inflammatory events, which unexpectedly exert a detrimental influence, as demonstrated in neurodegenerative diseases such as PD. When activated by pathological injury in the brain, microglial cells can rapidly change their morphological characteristics, increase their motility and phagocytic activity, and secrete cytokines that fight pathogens and promote tissue remodeling and repair [54]. Toll-like receptors (TLR) are a family of pattern-recognition receptors that recognize pathogen-related molecular patterns and misfolded proteins [56]. Microglia can recognize misfolded α-Syn by receptors such as TLR2 and TLR4 on the cell surface, so the binding of α-Syn to TLR2 and TLR4 can initiate related signaling pathways, such as NF-κB and the upregulation of proinflammatory cytokines and promote the transformation of anti-inflammatory microglial phenotype M2 into the proinflammatory microglial phenotype M1 [57]. The M2 phenotype of microglia can secrete anti-inflammatory factors IL-4, IL-10, and transforming growth factor-β (TGF-β), to significantly reduce and/or produce a large number of proinflammatory cytokines such as IL-1β, tumor necrosis factor-α (TNF-α) and reactive oxygen species (ROS) (Fig. 1) [57, 58]. Proinflammatory cytokines and ROS secreted by phenotypic M1 microglia further amplify neuroinflammatory responses in the brain and exacerbate the death of dopaminergic neurons [58].

PD and pyroptosis

Microglia are the main cell mediators of brain inflammation during the inflammatory response. Inflammatory responses mediated by inflammatory cytokines such as IL-1β secreted by microglia play an essential role in the development and progression of PD [59]. The major events that regulate the secretion of IL-1β by microglia are the activation of inflammasomes and a programmed inflammatory cell death process called pyroptosis [60]. Pyroptosis is a new type of programmed cell death related to an inflammatory response with similar characteristics but different mechanisms than apoptosis [61]. It is not regulated by the apoptosis-related protein caspase-3, but rather depends on the regulation of the inflammation-related protein caspase-1 [62]. The NLRP3 inflammasome plays an important role in pyroptosis, which is a critical component of the innate immune system that mediates caspase-1 activation and the secretion of proinflammatory cytokines [63]. Inflammasomes are associated with some autoimmune diseases and neurodegenerative diseases such as PD [64]. Activation of NLRP3 promotes the secretion of inflammatory factors such as IL-1β and IL-18 and induces pyroptosis, which enhances inflammatory effects by promoting the secretion of IL-1β and activation of inflammasomes [64, 65]. There is also a close correlation between pyroptosis and α-Syn aggregation. The aggregation of α-Syn can be released from damaged neurons into the extracellular space to recognize TLR receptors on microglia, which activates the NF-κB pathway and promotes the production of the IL-1β precursor protein [57, 66]. The aggregation of α-Syn in microglia activates NLRP3 inflammasomes and induces pyroptosis as a danger signal [67]. Inflammatory cytokines IL-1β and IL-18 secreted outside the cells further amplify the inflammatory response and damage dopaminergic neurons (Fig. 1).

In summary, the neuroimmune response induced by excessive or misfolded α-Syn is likely to be caused by microglia-mediated inflammation. The misfolded and deposited α-Syn can activate the adaptive immune response, which induces the pathological development of PD. This novel mechanism may explain the pathogenesis of PD and offer a new strategy for treatment.

Mechanisms and roles of microglia on T cells in PD

PD and T cell

In 1988, P L McGeer et al. identified a unique pattern of HLA-DR-positive microglia by double immunostaining for glial fibrillary acidic protein and HLA-DR in PD patient samples obtained after death, which indicated that the disease might be related to a neuroimmune response [68]. Studies have also shown a decrease in the number of T cells, including CD4 + and CD8 + T cells in the peripheral blood of PD patients, the high expression of MHC molecules in mononuclear cells, and the detection of a large number of activated T cells in the cerebrospinal fluid [69,70,71,72]. This phenomenon indicated that the peripheral immune components might enter the central nervous system in PD patients and that the ability of peripheral blood T cells to enter the brain may be the result of systemic inflammatory responses. Studies have shown that the integrity of the BBB in the ventral midbrain and deep cortex is damaged due to inflammation in PD patients and as a result, T cells can infiltrate into the brain (Fig. 1) [70, 73,74,75]. Studies have also shown that the levels of CD4 + and CD8 + T cells in the SN of the brain are significantly higher in PD patients than that in healthy people [76,77,78]. It is worth noting that T cells are mainly distributed near blood vessels and neuromelanin-positive dopaminergic neurons, which suggests that T cells are likely to interact with dopaminergic neurons through targeted migration [75, 79].

Microglia promote infiltration of CD8 + T cells

Roy et al. reported marked upregulation of RANTES (regulated on activation, normal T-cell expressed and secreted) and eotaxin, chemokines that are involved in T cell trafficking, in the serum of hemi-Parkinsonian monkeys [80]. The infiltration of CD8 + T cells around the hemi-Parkinsonian monkey’s parenchyma was significantly increased compared to that of the control group, which indicated that they were highly likely to migrate to the parenchyma by chemotaxis [80]. In addition, they discovered that in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) PD mouse model, the expression of RANTES and eotaxin was up-regulated in microglia cells of brain parenchyma, and confirmed that activation of the NF-κB signaling pathway is the most important regulatory pathway for inflammation in the upregulation of these two chemokines, because this pathway controlled the transcription and expression of proinflammatory factors such as RANTES and Eotaxin in many cells [81]. The NF-κB activation of microglia in the SN has been confirmed in MPTP-treated mice and monkeys and PD patients, and the role of NF-κB in the adaptive immune response of PD brain SN has been investigated [82,83,84]. These results suggest that activation of the NF-κB signaling pathway in microglia under pathological conditions of PD promotes the secretion of inflammatory chemokines such as RANTES and eotaxin, increasing the infiltration of CD8 + T cells into PD brain SN and the cytotoxic response to dopaminergic neurons (Fig. 1).

Microglia regulate CD4 + T cells

In 2017, two studies showed that autoreactive T cells could recognize specific α-Syn epitopes in PD patients [85, 86]. Researchers observed infiltration of CD4 + Th17 cells was also observed in an MPTP mouse model and that Th17 cells increased the death of dopaminergic neurons in MPTP-treated ventral midbrain dopaminergic neuron cultures [87, 88]. In the same study, the death of midbrain neurons derived from autologous induced pluripotent stem cells in PD patients may be closely related to CD4 + Th17 cells secreting IL-17, indicating that in addition to the neuronal susceptibility to CD8 + cytotoxic T lymphocytes, CD4 + T cells are also involved in the pathological process of PD [88]. The death of T lymphocyte-induced dopaminergic neurons was found to be mediated through the IL-17-IL-17R signaling pathway, which further activated the downstream NF-κB signaling pathway associated with cellular inflammatory and immune responses (Fig. 1) [88]. Activated microglia can be used as antigen-presenting cells (APCs) to present autoantigens such as α-Syn by MHC molecules [85]. The T cells activated by α-Syn misfolded proteins mainly involve CD4 + T cells, which proliferate and release large amounts of proinflammatory cytokines, leading to the apoptosis of dopaminergic neurons [87]. In addition to the activation of T cells by APCs, another clinical study of PD also showed that neuroinflammatory conditions such as secretion of inflammatory cytokines can trigger the expression of MHC-I molecules on the surface of dopaminergic neurons, making them vulnerable to attacks by CD8 + T cells, which also strongly supports the participation of the autoimmune response in PD patients [85].

In summary, CD4 + /CD8 + T cells expressing α-Syn were found in the brains of PD patients, where α-Syn could be recognized as an antigen by APCs, such as microglia or monocytes. The APCs can present antigens to CD4 + and CD8 + T cells, induce their activation, and trigger an autoimmune response in the brain, leading to the death of dopaminergic neurons.

Mechanisms and roles of T cells on microglial cells in PD

T cells regulate the activation of microglia

The regulation of T cells on microglia is also critical in the brain pathogenesis of PD patients. Some researchers have suggested that T cells are essential for the upregulation of MHC-II molecules in microglia, and this is an important process of α-Syn activation of microglia in the SN, leading to the loss of dopamine neurons [89,90,91]. David et al. found that α-Syn alone may not be sufficient to cause significant death of dopaminergic neurons in the SN, while T cells can accelerate the death of dopaminergic neurons by up-regulating the expression of MHC-II molecules in microglia [90]. This study found that MHC-II overexpression in vivo accelerated α-Syn-induced microglial activation, and MHC-II knockout prevented α-Syn-induced microglial activation, antigen presentation, and dopaminergic neuron degeneration. Meena et al. evaluated the effect of T cells on the neurodegeneration of PD by injecting the adeno-associated virus (AAV) encoding human wild-type α-Syn into the SN of T-cell-deficient rats [92]. The results showed no significant increase in microglial activation in the T-cell deficit group, whereas the rats without T-cell deficiency showed a significant increase in MHC-II positive microglial activation and significant loss of dopaminergic neurons. The presence of CD4 + and CD8 + T cells in the SN was also observed after MHC-II expression in microglia and loss of dopaminergic neurons [93]. The MHC is a group of cell surface proteins divided into two major classes of I and II molecules, that plays a fundamental role in adaptive immunity [94, 95]. Under normal conditions, MHC-I molecules are involved in endogenous antigen presentation and are commonly expressed in almost all cells, while MHC-II molecules are involved in exogenous antigen presentation and are mainly expressed by APCs, such as monocytes, macrophages, and dendritic cells [94]. Genome-wide association studies (GWAS) showed that PD was associated with the haplotype of the MHC-II gene, and the expression level of MHC-II was significantly increased in PD samples [96]. In addition, MHC-II positive microglia were detected in the SN of PD patients, and their expression level increased with the severity of the disease [90]. Activated microglia can express MHC-II molecules to further activate T cells, which in turn up-regulate the expression of MHC-II molecules in microglia to accelerate the activation process [91].

Homeostasis of Teffs and Tregs

It follows that T cells play a key role in the neuroimmune response of PD, but the regulatory effect of specific subtypes of T cells on microglia remains to be further explored. The T cells involved in the regulation of microglia in the neuroimmune system are classified as effector T cells (Teffs) and regulatory T cells (Tregs) and play a crucial role in maintaining neuroimmune homeostasis [97]. The balance of cellular immune function may control the rate of disease, and when this balance is disturbed, it can lead to disease. Studies have shown that CD4 + Teffs (such as Th1 and Th17) are involved in the process of neuron degeneration (such as in PD and MS) and maintain and accelerate the proinflammatory M1 phenotype of microglia by secreting proinflammatory phenotypic factors L17, IFN-γ, and granules B (Fig. 2) [45, 92, 98]. Similarly, in the MPTP model of PD, infiltrating T lymphocytes were found near the activated microglia and MHC-II was significantly overexpressed [92]. These MHC-II-overexpressed microglia further activated T lymphocytes through the antigen presentation process enhancing the immune response of Teffs to the neurodegenerative process.

Fig. 2
figure 2

Mechanisms and roles of T cells on microglia cells in PD. The damaged BBB allows T lymphocytes, including Tregs, to enter the brain in the early stages of the disease, and Tregs can also play a role in immune regulation (left of the illustration). The immunosuppressive properties of Tregs gradually decrease as the disease progresses (middle and late stage), breaking the homeostasis between Tregs and Teffs. Teffs, in particular, hasten the activation of microglia cells by secreting various proinflammatory factors, which hastens the onset of neurodegeneration (right of the illustration). The images in this Figure were downloaded from ProteinLounge.com with permission to use them

Tregs inhibit the activation of microglia

Tregs are naturally occurring subsets of CD4 + and CD25 + T lymphocytes [99]. In contrast to Teffs, anti-inflammatory phenotypes of cytokines secreted by Tregs have been shown to have neuroprotective effects. They are a major source of IL10 and TGF-β1 in vivo and are a major regulatory mechanism that controls innate and adaptive immune responses, so they have great potential to induce neuroprotection in PD [100]. Studies have shown that Tregs can inhibit microglial activation, promote neuronal survival in an MPTP-induced PD model, and are thought to inhibit the immune response through a variety of mechanisms [99, 100]. They also inhibit the functional metabolic pathway of Teffs through the release of anti-inflammatory factors such as IL-10 and TGF-β1. In addition to inhibiting the function and proliferation of Teffs, Tregs maintain anti-inflammatory microglia and astrocyte phenotypes by releasing IL-4, IL-10, and TGF-β (Fig. 2) [101, 102]. It has been shown that the transfer of CD4 + CD25 + Tregs to MPTP model mice with PD results in dose-dependent neuroprotective effects accompanied by inhibition of microglial activation, resulting in increased survival of dopaminergic neurons in the substantia nigra [99]. It has also been shown that Tregs can inhibit the function of M1-type microglia by inhibiting the activation of the NF-κB signaling pathway [91].

PD and T cell homeostasis

The disruption of Teff and Treg homeostasis plays a crucial role in the neuroimmune response mechanism of PD [91]. In the initial stage of the disease, the damaged BBB allows T lymphocytes, including Tregs, to enter the brain and Tregs can also play a role in immune regulation at this stage. However, with the progression of the disease, the immunosuppressive properties of Tregs gradually decrease, which breaks the homeostasis between Tregs and Teffs. The Teffs then accelerate the activation of microglial cells by secreting various proinflammatory factors and further accelerate the occurrence of neurodegeneration (Fig. 2) [92].

Summary of pathological mechanism

Taken together, the major factors that constitute the neuroimmune microenvironment of PD include a persistent inflammatory response, activated microglia, a balance disorder between Teffs and Tregs, and degeneration of dopaminergic neurons, which leads to the occurrence and development of PD (Fig. 3). The α-Syn is the main pathological marker of PD. Under normal physiological conditions, there is a dynamic balance between the production and degradation of α-Syn in the human body, but the dynamic balance is broken and α-Syn is misfolded due to the action of various genetically or environmentally controlled pathological factors. Besides the abnormal α-Syn accumulation in the brain, the abnormal α-Syn accumulation starting in the gut can also be transmitted to the brain through the vagus nerve. During the course of the disease, the α-Syn can spread to different brain regions in a prion-like fashion. Microglia are the first immune defense system of the human brain and one of the main cell types involved in the inflammatory response of the central nervous system. Microglia can recognize misfolded α-Syn through TLR2 and TLR4 receptors on the cell surface and further initiate related signaling pathways, such as activating the NF-κB signaling pathway and up-regulating the expression of proinflammatory cytokines, to promote the transformation of anti-inflammatory phenotype M2 into proinflammatory phenotype M1. The activation of the NF-κB signaling pathway in microglia under pathological conditions of PD promotes the secretion of inflammatory chemokines such as RANTES and eotaxin, increasing the infiltration of CD8 + T cells in PD brain SN and the cytotoxic response to dopaminergic neurons. In addition, α-Syn can be taken up as an antigen by microglia and then acts as APCs, which in turn provide antigens to CD4 + and CD8 + T cells and cause them to activate and trigger an autoimmune response through multiple pathways, eventually leading to the apoptosis of dopaminergic neurons. The regulation of microglia by T cells invading the brain of PD patients is also an important form of pathogenesis. In the initial stage of the disease, the impaired BBB allows T lymphocytes, including Tregs, to enter the brain. As the disease progresses, the homeostasis between Tregs and Teffs is disrupted, and Teffs accelerate the activation process by secreting various pro-inflammatory factors to up-regulate the expression of MHC-II molecules in microglia. Moreover, further activation of microglia also increases their effect on T cells, and the interaction between the two kinds of cells creates a vicious cycle that accelerates mitochondrial dysfunction and apoptosis in dopaminergic neurons. Inhibiting autoimmune responses through external intervention such as stem cell therapy and increasing the number of Tregs in the brain is likely to become an important strategy for PD treatment.

Fig. 3
figure 3

Neuroimmune microenvironment in PD. The neuroimmune microenvironment of PD is composed of several major factors, including a persistent inflammatory response (1), activated microglia (2), a balance disorder between Teffs and Tregs (3,4), and dopaminergic neurodegeneration (5), all of which contribute to the occurrence and progression of the disease through their interactive regulation. The images in this Figure were downloaded from ProteinLounge.com with permission to use them

Immunomodulatory therapies targeting α-Syn for PD

A-Syn is the primary pathological marker of PD and the most promising therapeutic target for PD immunotherapy. Misfolded α-Syn accumulates in microglia and dopaminergic neurons, which can activate various immune pathways. As a result, targeting α-Syn to prevent intracellular aggregation and propagation is an important strategy for the prevention and treatment of Parkinson's disease. The current PD treatment focuses primarily on α-Syn clearance and degradation, as well as immunoregulation therapy for autoimmunity.

A-Syn immunotherapy, both active and passive

Active and passive

A-Syn forms oligomers and fibrils depending on its biological properties and the situation. However, it is subject to posttranslational modifications such as acetylation, phosphorylation, and truncation [103, 104], and it also targets related immune mediators primarily via an immune pathway to accelerate dopaminergic neurodegeneration [105, 106]. Immunotherapy, including active and passive immunity, is based on the principle of specific antigen/antibody binding [107, 108]. Active immunity is the use of antigens to stimulate the body's production of antibodies rather than directly introducing antibodies from outside the body, which has a high resistance to subsequent infection, either through the disease pathogen itself or through immunization [109]. Data show that naturally occurring anti-α-Syn autoantibodies help with α-Syn clearance, reduce protein aggregation, and protect neurons [107, 110]. Active immunization against PD has been tested in animal models and early clinical trials in a large population, and it has proven to be less expensive than other treatments [107, 111]. Vaccinations do not require frequent injections, which can relieve financial and psychological stress on patients. Active immunity is more suitable for patients with early Parkinson's disease and high-risk groups because it relies on the use of specific antigens that can produce inflammatory response antibodies [112, 113]. Passive immunity is the specific immunity acquired by the body by passively receiving antibodies, sensitized lymphocytes, or their products. Antitoxin, placental globulin, antibacterial serum, and immune modulators are common biological products of artificial passive immunity [100, 114]. Passive immunization is thus appropriate for patients with moderate to severe PD.

A-Syn antibody preparations and clinical trials

Several studies have shown that an α-Syn transgenic mouse line, 83Vle/J, expressing human A53T variant α-Syn (the full-length, 140 amino acid isoform), can produce high-affinity antibodies and reduce α-Syn accumulation in dopaminergic neurons, thereby reducing neuronal damage [115, 116]. Furthermore, DNA vaccines can induce active immunity in the brain by inducing the production of antibodies that recognize misfolded α-Syn [117]. These peptide fragments, as well as the full-length recombinant α-Syn, were used to sensitize mouse dendritic cells, which were then intravenously delivered into transgenic mice expressing the human A53T variant of α-Syn, which could effectively produce specific antibodies and improve neuromotor function in vitro [116, 118]. Two short peptide vaccines (PD01A and PD03A) are being tested in clinical trials to assess the efficacy, tolerability, and safety of active immunization in Parkinson's disease patients (Table 1) [15, 16, 119]. Traditional antibodies, such as monoclonal (mAb) and polyclonal (pAb), have been at the forefront of biomedical research for diagnostic and analytical therapies against cancer, immune diseases, and infectious diseases [120]. They do, however, have limitations, such as stability over narrow pH and temperature ranges and the potential inability to access specific active sites on proteins [120]. Single-stranded antibodies have superior tissue penetration, the ability to cross the blood–brain barrier, the unique ability to bind to small cavities or clefts, high affinity and specificity, and high stability at extreme temperatures and pH [120, 121]. These advantages of single-stranded antibodies can improve their therapeutic effect and be used to treat PD [121]. A clinical trial of antibody preparation MEDI1341 was conducted to reduce extracellular α-Syn levels in healthy volunteers, and the study evaluated safety and tolerability (Table 1) [122]. MEDI1341 rapidly enters the central nervous system after intravenous injection into rats and cynomolgus monkeys, lowering free extracellular α-Syn levels in the interstitial fluid (ISF) and cerebrospinal fluid (CSF) compartments [123]. AstraZeneca and Takeda are collaborating to develop the antibody MEDI1341, which reportedly retains reduced immune effector function [122]. Clinical trials of BIIB054 and PRX002, which directly target the amino terminus of α-Syn and are highly selective for the aggregation form of α-Syn (Table 1), have also been conducted [17, 124, 125]. BIIB054 binds to α-Syn in PD and DLB tissue sections and extracts but not in unaffected brains and prevents dopamine transporter reduction induced by injection of preformed α-Syn fibrils into mouse brains [122]. This study found no evidence of clinical efficacy. Each of the three infusions resulted in significant reductions in free serum α-Syn levels within 1 h of PRX002 administration. PRX002 levels in serum and CSF increased in a dose-dependent manner [125]. These studies looked at the efficacy, safety, tolerability, pharmacokinetics, and pharmacodynamics of PD patients.

Table 1 Clinical trials of immunotherapy against α-Syn in PD

MSCs modulate α-Syn in PD: a multitarget disease-modifying therapies strategy

MSCs and neuroinflammatory disease

MSCs, as adult stem cells capable of paracrine, immune regulation, and multidirectional differentiation, have several advantages over embryonic stem cells, including a lower immune response, a lower risk of tumor formation, and no ethical concerns [126,127,128,129]. More research has been done on the relationship between MSCs and inflammatory processes since the discovery of their immunomodulatory properties. There is evidence that MSCs can migrate to the site of an inflammatory response via chemotaxis and have an immunomodulatory effect on specific chemotaxis recruitment responses, thereby reducing the inflammatory response in the damaged area and promoting tissue repair [130,131,132]. MSC-based cell therapies have been used to treat a variety of autoimmune-related neurodegenerative and neuroinflammatory diseases, including PD, Alzheimer's disease, multiple system atrophy, and amyotrophic lateral sclerosis [133,134,135,136]. The main pathogenic mechanism in PD is thought to be the loss of dopaminergic neurons, and most researchers believe that the therapeutic effect is due to MSCs' cell replacement ability. MSC research has shifted in recent years from cell replacement to multitarget therapy such as paracrine and immune regulation (Table 2).

Table 2 multitarget disease-modifying therapies of MSCs modulates α-Syn in PD

Toxin-based animal models

PD is a multifactorial disease caused by genetic and environmental factors. Toxin-based animal models (such as 6-OHDA and MPTP) have primarily aided in the development of treatments for motor symptoms. Toxin-based models have advantages in that the symptoms are clear, the experiment period is short, they can be applied to a wide range of animal species, they show prodromal symptoms, and they will be used in future models [137]. According to Roy A et al., T-cell migration-related cytokines RANTES and eotaxin were up-regulated in microglia in the brain parenchyma of MPTP monkey models, while CD8 + T cell infiltration around the deep brain parenchyma of PD monkeys was significantly increased [80]. This study also showed that MPTP animal models could simulate some pathological processes in PD patients.

Transgenic animal models

The α-Syn gene, the causative gene of familial PD, is one of the most important genetic factors in PD patients, and many α-Syn transgenic animal models have been created, contributing to the elucidation of PD pathology [137, 138]. The α-Syn transgenic model can replicate α-Syn aggregation and show slowly progressive changes, similar to human PD. Furthermore, injected α-Syn fibrils have been shown in mouse experiments to spread throughout the brain [139]. The propagation model created by exogenously administered α-Syn fibrils is still developing, but obtaining a phenotype is relatively simple, and the experiment takes less time than the transgenic model [139, 140]. The fact that this model can be applied to primates and other animals is a huge plus.

According to one study, exosomes isolated from MSCs can significantly improve the motor, learning, and memory abilities of the progressive in α-Syn A53T transgenic mice model, and the mechanism may be related to changes in phospholipid composition and cholesterol metabolism in hippocampal neurons [141]. Currently, the use of MSCs in transgenic mouse models is relatively rare, with most studies focusing on toxin-based models. As a result, we will investigate the role and mechanism of MSC intervention in MPTP toxicity and α-Syn-induced cell models.

Modulating microglia activation

Microglia cells are the most active immune cells in brain tissue, and their mediated inflammatory response is linked to the pathogenesis of many immune-related nervous system diseases. Changes in the extracellular microenvironment can cause microglia to polarize into two cell phenotypes, M1 and M2. Misfolded α-Syn, though toxic to dopaminergic neurons, can activate microglia cells, causing them to polarize to the M1 phenotype, amplifying the neuroinflammatory response and secreting numerous proinflammatory factors [144, 145]. Because the phenotype mediated by α-Syn is involved in the pathogenesis of PD, external intervention to promote the polarization of microglia toward the M2 phenotype has important clinical value in the neuroinflammatory response [146, 147]. Lee et al. demonstrated that MSCs could induce M2 phenotype microglia cell polarization, significantly improve phagocytic clearance of α-Syn in microglial cells, and have a significant neuroprotective effect in α-Syn-rich cells and animal models [26, 28, 146]. In the same study, the activated recombinant α-Syn protein was injected unilaterally into the right neocortex of mice. The findings confirmed that MSCs could promote the polarization of M2 phenotype microglia and reduce α-Syn aggregation in cortical areas by activating IL-4-related STAT signaling pathways in microglia cells and α-Syn inoculated IL-4 KO mice [146]. Bidirectional studies using siIL-4 MSCs in α-Syn mice and MSCs in IL-4R-KO mice confirmed that they could counteract α-Syn phagocytosis of microglia cells by promoting M1 polarization [146]. They discovered that cerebrospinal fluid (CSF) from MSC-transplanted multiple system atrophy patients induced microglia M2 polarization and had a prosurvival effect in α-Syn-treated BV2 cells via enhanced clearance of α-Syn [146]. Another study found that MSCs could regulate microglial cell activation and reduce the inflammatory cascade reaction of activated microglia cells via TNF- stimulated gene/protein 6 (TSG-6), implying that MSCs and TSG-6 could be effective ways to treat neuroinflammatory diseases associated with microglial cell activation [148].

Enhancing autophagy

Autophagy is the primary pathway for the degradation of harmful aggregation proteins in a variety of neurodegenerative diseases. Autophagy has been shown to play an important role in the pathogenesis of PD, and autophagy disorders can result in abnormal α-Syn aggregation, which can be cleared by increased autophagy [27, 149]. This suggests that autophagy regulation could be used to treat PD. The PD models exhibited increased autophagy and neuronal apoptosis, owing to the failure of most autophagosomes to fuse with lysosomes as a result of α-Syn misfolding and the resulting neuroinflammatory response [150,151,152,153]. MSCs have been shown in studies to significantly increase autophagosome formation and reduce α-Syn aggregation after intervention in PD models, and the neuroprotective effect of MSCs is largely dependent on lysosomal activity mediated by autophagosome formation [154, 155]. Park et al. used human bone marrow-derived MSCs to interfere with an MPTP-treated neuron cell model, which significantly reduced neuron apoptosis and abnormal α-Syn expression while increasing the number of LC3-positive autophagosomes (Fig. 4) [154]. MSCs derived from human bone marrow were injected into the tail vein of MPTP-treated PD animal models, which can also significantly enhance the maturation of late autophagic vacuoles and fuse with lysosomes to form autophagosomes [154]. These findings suggest that MSC therapy could significantly improve autophagosome formation and α-Syn clearance in PD models.

Fig. 4
figure 4

Mechanisms of MSC multitarget immunotherapy in PD. Through their paracrine effect, MSCs secrete a large number of immunomodulatory cytokines. MSCs can promote M2 phenotype microglia polarization and reduce α-Syn aggregation in cortical areas by activating IL-4-related STAT signaling pathways (1). MSCs can significantly increase the formation of LC3-positive autophagosomes and decrease α-Syn aggregation (2) by activating the autophagy signaling pathway. MSCs inhibit extracellular α-Syn CME by regulating clathrin and EEA1, as well as the interaction between α-Syn and nNMDA receptors by releasing Gal-1, which completely binds NMDA receptors with α-Syn (3, 4). MSCs can eliminate abnormal α-Syn expression and accelerate its degradation via secretory small molecule cleavage, such as MMP-2 (5). We hypothesized that hypoxia preconditioning MSCs would prevent pyroptosis in microglia cells by down-regulating the NLRP3 inflammasome via the HIF-1 signaling pathway (6). MSCs can significantly induce M2 phenotype microglia cell polarization and have a significant dopaminergic neuronal protective effect by attenuating inflammation amplification (7). MSCs can significantly induce M2 phenotype microglia cell polarization and have a significant dopaminergic neuronal protective effect by attenuating the antigen presentation process and T cell chemokine secretion (8)

Mediating endocytosis

Extracellular α-Syn aggregates' prion-like behavior is important in the pathogenesis and progression of α-Synucleinopathies [156,157,158]. Although the precise mechanism of α-Syn intercellular transmission is unknown, receptor-mediated endocytosis is likely to be an important route for α-Syn into cells. MSCs have been shown to inhibit extracellular α-Syn clathrin-mediated endocytosis (CME) by modulating interactions with n-methyl-d-aspartic acid (NMDA) receptors [26]. MSCs can inhibit extracellular α-Syn CME by regulating clathrin and early endoplasmic reticulum antigen 1 (EEA1), as well as the interaction between α-Syn and the NMDA receptor in α-Syn aggregated cells (Fig. 4) [26]. MSCs were also found to reduce α-Syn endocytosis by releasing galectin-1 (Gal-1) to competitively bind NMDA receptors with α-Syn [26].

Protease secretion

Because protease can cut abnormal α-Syn aggregates, using an exogenous protease's cutting function may be able to clear abnormal α-Syn aggregation in PD. MSCs have been shown to secrete molecular substances into the nervous system microenvironment to promote proteolysis. MMP-2 is a soluble factor derived from MSCs that can participate in the hydrolysis of α-Syn proteins [28, 159]. MMP-2 molecules can disrupt newly formed amyloid deposition, reducing α-Syn insolubility and oligomeric levels significantly (Fig. 4) [28]. MSCs can thus eliminate abnormal α-Syn expression and accelerate its degradation via their small secretory molecule.

The hypothesis of inhibiting pyroptosis

MSC therapy for PD associated with microglia pyroptosis has not yet been studied. In vitro, hypoxia-preconditioned olfactory mucosal mesenchymal stem cells (OM-MSC) inhibit pyroptotic death of microglial cells in response to cerebral ischemia/reperfusion injury by activating hypoxia-inducible factor-1 (HIF-1) [160]. The activation of the NLRP3 inflammasome in BV2 microglia cells exposed to ischemia/reperfusion injury was investigated in this study, and it was discovered that the expression of ASC, caspase 1, caspase 8, and GSDMD proteins was increased after ischemia/reperfusion injury. Furthermore, IL-1 and 1L-18 levels increased following injury. It was also shown that co-culture with hypoxia preconditioned OM-MSCs significantly reduced pyroptotic cell death in microglia following cerebral ischemia/reperfusion injury compared to normal OM-MSCs. In addition, α-Syn aggregation in microglia activates NLRP3 inflammasomes and induces pyroptosis as a danger signal, causing microglia to switch from anti-inflammatory phenotype M2 to pro-inflammatory phenotype M1 [67]. IL-1 and IL-18 as two inflammatory cytokines produced extracellularly increase the inflammatory response and destroy dopaminergic neurons. Furthermore, hypoxia-preconditioned OM-MSCs dramatically reduce IL-1 and IL-18 production in microglia, indicating that OM-MSCs may prevent pyroptosis in microglia. It was hypothesized that hypoxia preconditioning of MSCs may prevent pyroptosis of microglia cells in neurodegenerative diseases associated with neuroinflammatory responses, such as PD (Fig. 4).

Summary of multitarget disease-modifying therapies

Taken together, dopaminergic neuron degeneration is caused by the interaction of α-Syn, microglia, and T cells during the course of PD. As a result, if one or more of these links can be targeted to slow disease progression, it will be an important strategy for PD prevention and treatment. In summary, the use of MSCs can accelerate the clearance and degradation of α-Syn via multitarget disease-modifying therapies mechanisms such as modulating microglia activation, enhancing autophagy, mediating endocytosis, protease secretion, and possibly preventing pyroptosis of microglia. Furthermore, one study found that MSCs inhibit the differentiation of CD4 + T cells into Th17 cells isolated from PD patients and that this suppressive effect was primarily associated with an increase in functional CD25 + Foxp3 + Treg cells and IL-10 secretion [161]. Furthermore, studies have shown that MSCs can be induced to differentiate into dopaminergic neurons and improve neural function by replacing lost neurons in the brain of people with PD. MSCs as cell-based therapy candidates in PD have a broader scope than simple cell type replacement because they can be used as a cellular system for the detoxification of reactive oxygen species (ROS) as well as a supplier of neurotrophic factors via paracrine effect to protect the mitochondrial function of dopaminergic neurons [162]. Based on progress in understanding MSCs' multitarget disease-modifying therapies effect, we believe MSCs could be used as an important strategy for therapy or to prevent the onset of PD in at-risk individuals, as well as to slow the progression of the disease.

Major challenges of MSC-based cell therapy

Allogeneic and autologous MSCs

Although MSCs have tremendous potential and opportunities in the treatment of immune diseases such as PD, their clinical application faces numerous challenges. The differences in cell sources and amplification schemes, as well as the decreased survival rate, paracrine effect, and homing ability caused by the local adverse microenvironment following cell transplantation, limit MSCs' therapeutic effect. MSCs derived from allogenic sources have received the most attention and application over the last few decades. Despite numerous reports describing MSC immune privilege or low immunogenicity, other studies have shown that allogeneic MSCs enhance inflammation and are subsequently cleared in response to rejection by donor-specific antibody levels, suggesting that this upregulation may be the reason for allogeneic MSC treatment failure [163, 164]. Interestingly, one study found that the immunogenicity of MSCs depended on whether they differentiated in vivo, with undifferentiated MSCs having low immunogenicity, while MSCs after differentiation had significantly higher immunogenicity [165]. These factors also reduce MSCs' therapeutic effect to some extent [163, 165]. Although allogeneic MSCs may be immunogenic, their potent immunomodulatory effects have opened up new avenues for immunomodulatory inflammatory diseases and attracted researchers in related fields. Researchers discovered in 2010 that OM-MSCs isolated from human nasal mucosa were a better source of MSCs [166]. Since 2014, our research team has been studying autologous human OM-MSCs and has developed a complete culture system. These cells have a higher rate of proliferation and a shorter passage time. OM-MSCs are widely distributed in the nasal cavity, are easily accessible, exhibit no immune rejection, have few or no associated ethical issues [127, 167], and have the added benefit of retaining biological activity that does not change with age [168]. As previously stated, nasal mucosa develops from embryonic ectoderm and can directly differentiate into neurons under the right conditions. According to chromosome karyotype analysis and tumor gene analysis, there is no genetic variation after substantial in vitro passages [169]. It is unknown, however, whether the success of autologous MSC transplantation is impacted by pathologic genetic background since there have been no reports of clinical studies using autologous MSC transplantation for the treatment of neurodegenerative diseases. Recently, we confirmed that OM-MSCs transplantation could alleviate the symptoms of Alzheimer's disease in APPswe/PS1dE9 mice and promote Aβ clearance through immunomodulation, thus demonstrating the great potential and social value of OM-MSC treatment for Alzheimer's disease patients. What's more, our findings confirmed that OM-MSCs could differentiate in multiple directions, including neurons, bone tissue, and adipose tissue [170]. Thus, the benefits of OM-MSCs make them an ideal source of cells for treating neuroimmune diseases such as PD.

Challenges and improvements for MSCs

The cell survival rate, number of homing cells, and immunomodulatory effects of MSCs after implantation in vivo are critical to the success of MSCs therapy. Excessive inflammatory response, oxidative stress, hypoxia, and other negative factors in the microenvironment at the site of injury of the inflammatory immune disease may be the most important factors limiting MSC survival and efficacy. During in vitro amplification, these MSCs were cultured in 21% oxygen and 10–20% serum concentration, but after implantation in disease models or patients, they were exposed to hypoxic or ischemic microenvironments. In this case, MSCs rely solely on anaerobic glycolysis activation to obtain weak energy and may undergo self-apoptosis [171]. Many studies have shown that pretreatment with hypoxia, hypertrophic medium, cytokines, or chemicals can improve MSC survival, homing ability, and paracrine effect after transplantation [172, 173]. The primary goal of MSC pretreatment is to allow them to adapt to the harsh local microenvironment ahead of time while also increasing their survival rate, paracrine, immune regulation, and differentiation functions. Previous research has shown that a hypoxic microenvironment can promote OM-MSC proliferation and differentiation into dopaminergic neurons [128]. In vitro, hypoxia-preconditioned OM-MSC inhibited pyroptotic death of microglial cells in response to cerebral ischemia–reperfusion insult by activating hypoxia-inducible factor-1 [160]. Hypoxic pretreatment has also been shown in studies to improve MSC migration and homing ability and increase the number of damaged local MSCs [174, 175]. Furthermore, the immunomodulatory effects of MSCs are mediated by a cascade of inflammatory cytokines at the site of implantation [176], and preconditioning MSCs with inflammatory cytokines can increase the expression of intercellular adhesion molecules, inducible oxide synthase (iNOS), and chemokines while inhibiting T cell proliferation [177, 178]. The optimal culture scheme for MSCs to improve their survival rate, homing ability, and paracrine effect is a promising field of research that will address some of the challenges associated with MSC therapy. Finally, while MSC therapy has demonstrated pre-clinical success in treating various diseases, many other issues have been discovered during clinical trials. There is currently no international standard for the clinical application of MSCs, including cell dose, administration interval time, culture conditions, and route of administration. More research on the effects of the factors above and the biological characteristics of MSCs derived from various sources is required to find the best treatment for the various diseases and the potential for MSC-based therapies.

Conclusions

In summary, α-Syn is the primary pathological marker of Parkinson's disease, and it can accumulate in the SN, triggering a neuroinflammatory response by activating microglia. This marker can further activate the neuroimmune response of dopaminergic neurons, which is mediated by reactive T cells via antigen presentation. The main components of the PD neuroimmune microenvironment are a persistent inflammatory response, activated microglia, a balance disorder between Teffs and Tregs, and dopaminergic neurodegeneration, which interact to cause the occurrence and progression of PD. MSCs can help to reduce the burden of toxic aα-Syn by promoting the M2 phenotype of microglia, increasing autophagy, increasing proteolysis of α-Syn aggregates, and inhibiting α-Syn intercellular transmission, allowing treatment via multitarget dominant immune regulation. Abnormal protein expression can be found in a variety of diseases with varying phenotypes, implying that abnormal α-Syn expression is a by-product of other major pathogenic pathways or immunomodulatory processes. Multitarget disease-modifying therapies involving neurodegenerative disease strategies are critical for the clinical efficacy of diseases characterized by abnormal protein expression, such as α-Syn, and the use of MSCs could be the most promising candidate for future treatment strategies.

Availability of data and materials

Not applicable.

Abbreviations

PD:

Parkinson’s disease

SN:

Substantia nigra

A-syn:

Alpha-synuclein

MSCs:

Mesenchymal stem cells

BBB:

Blood–brain barrier

UPS:

Ubiquitin proteasome system

ALP:

Autosomal system

TLR:

Toll-like receptors

TGF-β:

Transforming growth factor-β

TNF-α:

Tumor necrosis factor-α

ROS:

Reactive oxygen species

NLRP3:

NLR family, pyrin domain-containing protein 3

ASC:

Apoptosis-related speck protein containing the caspase activation recruitment domain

MHC:

Major histocompatibility complex

RANTES:

Regulated on activation, normal T-cell expressed and secreted

MPTP:

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine

APCs:

Antigen-presenting cells

AD:

Alzheimer’s disease

MSA:

Multiple system atrophy

ALS:

Amyotrophic lateral sclerosis

CSF:

Cerebrospinal fluid

TSG-6:

TNF-α-stimulated gene/protein 6

LC3:

Microtubule-associated protein 1 light chain 3

CME:

Clathrin-mediated endocytosis

EEA1:

Early endoplasmic reticulum antigen 1

NMDA:

N-methyl-d-aspartic acid

Gal-1:

Galectin-1

MMP-2:

Matrix metalloproteinase-2

OM-MSCs:

Olfactory mucosal mesenchymal stem cells

HIF-1α:

Hypoxia-inducible factor-1α

References

  1. Zeng XS, Geng WS, Jia JJ, Chen L, Zhang PP. Cellular and molecular basis of neurodegeneration in Parkinson Disease. Front Aging Neurosci. 2018 ;10:109.

  2. Simuni T, Uribe L, Cho HR, et al. Clinical and dopamine transporter imaging characteristics of non-manifest LRRK2 and GBA mutation carriers in the Parkinson’s Progression Markers Initiative (PPMI): a cross-sectional study. Lancet Neurol. 2020;19(1):71–80.

    Article  CAS  PubMed  Google Scholar 

  3. Olanow CW, Factor SA, Espay AJ, et al. Apomorphine sublingual film for off episodes in Parkinson’s disease: a randomised, double-blind, placebo-controlled phase 3 study. Lancet Neurol. 2020;19(2):135–44.

    Article  CAS  PubMed  Google Scholar 

  4. Shahnawaz M, Mukherjee A, Pritzkow S, et al. Discriminating αα-Synuclein strains in Parkinson’s disease and multiple system atrophy. Nature. 2020;578(7794):273–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gallea JI, Ambroggio EE, Vilcaes AA, James NG, Jameson DM, Celej MS. Amyloid oligomerization of the Parkinson’s disease-related protein αα-Synuclein impacts on its curvature-membrane sensitivity. J Neurochem. 2018;147(4):541–56.

    Article  CAS  PubMed  Google Scholar 

  6. Ho PW, Leung CT, Liu H, et al. Age-dependent accumulation of oligomeric SNCA/αα-Synuclein from impaired degradation in mutant LRRK2 knock-in mouse model of Parkinson disease: role for therapeutic activation of chaperone-mediated autophagy (CMA). Autophagy. 2020;16(2):347–70.

    Article  CAS  PubMed  Google Scholar 

  7. Espay AJ, Vizcarra JA, Marsili L, et al. Revisiting protein aggregation as pathogenic in sporadic Parkinson and Alzheimer diseases. Neurology. 2019;92(7):329–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pang SY, Ho PW, Liu HF, et al. The interplay of aging, genetics and environmental factors in the pathogenesis of Parkinson’s disease. Transl Neurodegener. 2019;8:23.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173(4):649–65.

    Article  CAS  PubMed  Google Scholar 

  10. Culmsee C, Michels S, Scheu S, Arolt V, Dannlowski U, Alferink J. Mitochondria, microglia, and the immune system-how are they linked in affective disorders. Front Psychiatry. 2018;9:739.

    Article  PubMed  Google Scholar 

  11. Zhang W, Wang T, Pei Z, et al. Aggregated alphaα-Synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J. 2005;19(6):533–42.

    Article  CAS  PubMed  Google Scholar 

  12. La Vitola P, Balducci C, Baroni M, et al. Peripheral inflammation exacerbates αα-Synuclein toxicity and neuropathology in Parkinson's models. Neuropathol Appl Neurobiol. 2020.

  13. Bhattacharyya KB. The story of Levodopa: a long and arduous journey. Ann Indian Acad Neurol. 2022;25(1):124–30.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Przytuła F, Dulski J, Sobstyl M, Sławek J. Battery for deep brain stimulation depletion in Parkinson’s Disease and dystonia patients—a systematic review. Neurol Neurochir Pol. 2021;55(4):346–50.

    Article  PubMed  Google Scholar 

  15. Schneeberger A, Mandler M, Mattner F, Schmidt W. Vaccination for Parkinson’s disease. Parkinsonism Relat Disord. 2012;18(Suppl 1):S11–3.

    Article  PubMed  Google Scholar 

  16. Braczynski AK, Schulz JB, Bach JP. Vaccination strategies in tauopathies and synucleinopathies. J Neurochem. 2017;143(5):467–88.

    Article  CAS  PubMed  Google Scholar 

  17. Brys M, Fanning L, Hung S, et al. Randomized phase I clinical trial of anti-αα-Synuclein antibody BIIB054. Mov Disord. 2019;34(8):1154–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Danielyan L, Beer-Hammer S, Stolzing A, et al. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer’s and Parkinson’s disease. Cell Transplant. 2014;23(Suppl. 1):S123–39.

    Article  PubMed  Google Scholar 

  19. Yao Y, Huang C, Gu P, Wen T. Combined MSC-secreted factors and neural stem cell transplantation promote functional recovery of PD rats. Cell Transplant. 2016;25(6):1101–13.

    Article  PubMed  Google Scholar 

  20. Venkatesh K, Sen D. Mesenchymal stem cells as a source of dopaminergic neurons: a potential cell-based therapy for Parkinson’s disease. Curr Stem Cell Res Ther. 2017;12(4):326–47.

    Article  CAS  PubMed  Google Scholar 

  21. Shall G, Menosky M, Decker S, et al. Effects of passage number and differentiation protocol on the generation of dopaminergic neurons from rat bone marrow-derived mesenchymal stem cells. Int J Mol Sci. 2018;19(3).

  22. Shiying L, Xinhui Q, Guanghua J, et al. Puerarin promoted proliferation and differentiation of dopamine-producing cells in Parkinson’s animal models. Biomed Pharmacother. 2018;106:1236–42.

    Article  CAS  PubMed  Google Scholar 

  23. Chen Y, Shen J, Ke K, Gu X. Clinical potential and current progress of mesenchymal stem cells for Parkinson’s disease: a systematic review. Neurol Sci. 2020;41(5):1051–61.

    Article  PubMed  Google Scholar 

  24. Hong C-G, Chen M-L, Duan R, et al. Transplantation of Nasal olfactory mucosa mesenchymal stem cells benefits Alzheimer’s disease. Mol Neurobiol. 2022;59(12):7323–36.

    Article  CAS  PubMed  Google Scholar 

  25. Park HJ, Oh SH, Kim HN, Jung YJ, Lee PH. Mesenchymal stem cells enhance αα-Synuclein clearance via M2 microglia polarization in experimental and human parkinsonian disorder. Acta Neuropathol. 2016;132(5):685–701.

    Article  CAS  PubMed  Google Scholar 

  26. Oh SH, Kim HN, Park HJ, et al. Mesenchymal stem cells inhibit transmission of αα-Synuclein by modulating Clathrin-mediated endocytosis in a Parkinsonian model. Cell Rep. 2016;14(4):835–49.

    Article  CAS  PubMed  Google Scholar 

  27. Liang Y, Zhou T, Chen Y, et al. Rifampicin inhibits rotenone-induced microglial inflammation via enhancement of autophagy. Neurotoxicology. 2017;63:137–45.

    Article  CAS  PubMed  Google Scholar 

  28. Oh SH, Kim HN, Park HJ, Shin JY, Kim DY, Lee PH. The cleavage effect of mesenchymal stem cell and its derived matrix metalloproteinase-2 on extracellular αα-Synuclein aggregates in Parkinsonian models. Stem Cells Transl Med. 2017;6(3):949–61.

    Article  CAS  PubMed  Google Scholar 

  29. Yadav K, Yadav A, Vashistha P, Pandey VP, Dwivedi UN. Protein misfolding diseases and therapeutic approaches. Curr Prot Pept Sci. 2019;20(12):1226–45.

    Article  CAS  Google Scholar 

  30. Ma J, Gao J, Wang J, Xie A. Prion-like mechanisms in Parkinson’s disease. Front Neurosci. 2019;13:552.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Caputi V, Giron MC. Microbiome-gut-brain axis and toll-like receptors in Parkinson's disease. Int J Mol Sci. 2018;19(6).

  32. Prigent A, Alvarez-Fischer D, Callebert J, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Investig. 2009;119(1):182–92.

    PubMed  Google Scholar 

  33. Westhoff D, Witlox J, Koenderman L, et al. Preoperative cerebrospinal fluid cytokine levels and the risk of postoperative delirium in elderly hip fracture patients. J Neuroinflammation. 2013;10:122.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Raj T, Rothamel K, Mostafavi S, et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science. 2014;344(6183):519–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li Y, Zhou D, Ren Y, et al. Mir223 restrains autophagy and promotes CNS inflammation by targeting ATG16L1. Autophagy. 2019;15(3):478–92.

    Article  CAS  PubMed  Google Scholar 

  36. Cooper MD, Miller J. Discovery of 2 distinctive lineages of lymphocytes, T cells and B cells, as the basis of the adaptive immune system and immunologic function: 2019 Albert Lasker Basic Medical Research Award. JAMA. 2019;322(13):1247–8.

    Article  CAS  PubMed  Google Scholar 

  37. Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunol Rev. 2020;294(1):164–76.

    Article  CAS  PubMed  Google Scholar 

  38. Yin L, Scott-Browne J, Kappler JW, Gapin L, Marrack P. T cells and their eons-old obsession with MHC. Immunol Rev. 2012;250(1):49–60.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Brummelman J, Pilipow K, Lugli E. The single-cell phenotypic identity of human CD8+ and CD4+ T cells. Int Rev Cell Mol Biol. 2018;341:63–124.

    Article  CAS  PubMed  Google Scholar 

  40. Bradstreet JJ, Ruggiero M, Pacini S. Commentary: structural and functional features of central nervous system lymphatic vessels. Front Neurosci. 2015;9:485.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Louveau A, Smirnov I, Keyes TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Louveau A, Smirnov I, Keyes TJ, et al. Corrigendum: Structural and functional features of central nervous system lymphatic vessels. Nature. 2016;533(7602):278.

    Article  CAS  PubMed  Google Scholar 

  43. Sanchez-Guajardo V, Barnum CJ, Tansey MG, Romero-Ramos M. Neuroimmunological processes in Parkinson’s disease and their relation to αα-Synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro. 2013;5(2):113–39.

    Article  CAS  PubMed  Google Scholar 

  44. Csencsits-Smith K, Suescun J, Li K, Luo S, Bick DL, Schiess M. Serum lymphocyte-associated cytokine concentrations change more rapidly over time in multiple system atrophy compared to Parkinson disease. NeuroImmunoModulation. 2016;23(5–6):301–8.

    Article  CAS  PubMed  Google Scholar 

  45. Elgueta D, Contreras F, Prado C, et al. Dopamine receptor D3 expression is altered in CD4+ T-Cells from Parkinson’s Disease patients and its pharmacologic inhibition attenuates the motor impairment in a mouse model. Front Immunol. 2019;10:981.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Elfil M, Kamel S, Kandil M, Koo BB, Schaefer SM. Implications of the Gut Microbiome in Parkinson’s Disease. Mov Disord. 2020;35(6):921–33.

    Article  CAS  PubMed  Google Scholar 

  47. Braak H, Rüb U, Gai WP, Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna). 2003;110(5):517–36.

    Article  CAS  PubMed  Google Scholar 

  48. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alphaα-Synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.

    Article  CAS  PubMed  Google Scholar 

  49. Lockhart PJ, O’Farell C, Farrer MJ, West AB. Identification of a novel gene linked to parkin via a bi-directional promoter. J Mol Biol. 2003;326(1):11–9.

    Article  PubMed  Google Scholar 

  50. Lowe J, Layfield R, Cavey JR. Role of ubiquitin-mediated proteolysis in the pathogenesis of neurodegenerative disorders. Ageing Res Rev. 2003;2(4):343–56.

    Article  PubMed  Google Scholar 

  51. Soda M, Abe K, Murakami T, Shoji M, Imai Y, Takahashi R. A product of the human gene adjacent to parkin is a component of Lewy bodies and suppresses Panel receptor-induced cell death. J Biol Chem. 2003;278(51):51901–10.

    Article  PubMed  Google Scholar 

  52. McKeon JE, Sha D, Li L, Chin L. Parkin-mediated K63-polyubiquitination targets ubiquitin C-terminal hydrolase L1 for degradation by the autophagy-lysosome system. Cell Mol Life Sci CMLS. 2015;72(9):1811–24.

    Article  CAS  PubMed  Google Scholar 

  53. Huo H, Yan C, Liu Y, Chu Y, Yang C, Zhang T. Ubiquitin C-Terminal Hydrolase L1 regulates autophagy by inhibiting autophagosome formation through its deubiquitinating enzyme activity. Biochem Biophys Res Commun. 2018;497(2):726–33.

    Article  PubMed  Google Scholar 

  54. McCarthy RC, Sosa JC, Gardeck AM, Baez AS, Lee CH, Wessling-Resnick M. Inflammation-induced iron transport and metabolism by brain microglia. J Biol Chem. 2018;293(20):7853–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tanaka M, Sackett S, Zhang Y. Endocannabinoid modulation of microglial phenotypes in neuropathology. Front Neurol. 2020;11:87.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kouli A, Horne CB, Williams-Gray CH. Toll-like receptors and their therapeutic potential in Parkinson’s disease and αα-Synucleinopathies. Brain Behav Immun. 2019;81:41–51.

    Article  CAS  PubMed  Google Scholar 

  57. Rocha Sobrinho H, Silva D, Gomides LF, Dorta ML, Oliveira M, Ribeiro-Dias F. TLR4 and TLR2 activation is differentially associated with age during Parkinson’s disease. Immunol Invest. 2018;47(1):71–88.

    Article  PubMed  Google Scholar 

  58. da Silva DJ, Borges AF, Souza PO, et al. Decreased toll-like receptor 2 and toll-like receptor 7/8-induced cytokines in Parkinson’s disease patients. NeuroImmunoModulation. 2016;23(1):58–66.

    Article  PubMed  Google Scholar 

  59. Cheng J, Liao Y, Dong Y, et al. Microglial autophagy defect causes Parkinson disease-like symptoms by accelerating inflammasome activation in mice. Autophagy. 2020;1–13.

  60. Wang X, Teng H, Dai W, Wang J, Li C, Wang B. Celastrol inhibits microglial pyroptosis and attenuates inflammatory reaction in acute spinal cord injury rats. Int Immunopharmacol. 2019;66:215–23.

    Article  PubMed  Google Scholar 

  61. Aderem A, Rajan JV, Miao EA. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243:206–14.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Taabazuing CY, Okondo MC, Bachovchin DA. Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages. Cell Chem Biol. 2017;24(4):507–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci. 2019;20(13).

  64. Gouravani M, Khalili N, Razi S, Keshavarz-Fathi M, Khalili N, Rezaei N. The NLRP3 inflammasome: a therapeutic target for inflammation-associated cancers. Expert Rev Clin Immunol. 2020;16(2):175–87.

    Article  CAS  PubMed  Google Scholar 

  65. Zhang X, Ma X, Zheng X, Pan L. NLRP3 inflammasome activation in liver cirrhotic patients. Biochem Biophys Res Commun. 2018;505(1):40–4.

    Article  PubMed  Google Scholar 

  66. Aslanidis A, Karlstetter M, Scholz R, et al. Activated microglia/macrophage whey acidic protein (AMWAP) inhibits NFκB signaling and induces a neuroprotective phenotype in microglia. J Neuroinflammation. 2015;12:77.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Chen Y, Wen L, Zhang Q, et al. NLRP3 inflammasome activation in the thymus of MPTP-induced Parkinsonian mouse model. Toxicol Lett. 2018;288:1–8.

    Article  PubMed  Google Scholar 

  68. McGeer PL, Itagaki S, McGeer EG. Expression of the histocompatibility glycoprotein HLA-DR in neurological disease. Acta Neuropathol. 1988;76(6):550–7.

    Article  CAS  PubMed  Google Scholar 

  69. Alphaα-Synuclein induces apoptosis by altered expression in human peripheral lymphocyte in Parkinson's disease.

  70. Peripheral blood lymphocyte phenotypes in Alzheimer and Parkinson's diseases.

  71. Baba Y, Kuroiwa A, Uitti RJ, Wszolek ZK, Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat Disord. 2005;11(8):493–8.

    Article  PubMed  Google Scholar 

  72. Green H, Zhang X, Tiklova K, et al. Alterations of p11 in brain tissue and peripheral blood leukocytes in Parkinson’s disease. Proc Natl Acad Sci USA. 2017;114(10):2735–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. González H, Elgueta D, Montoya A, Pacheco R. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J Neuroimmunol. 2014;274(1/2):1–13.

    Article  PubMed  Google Scholar 

  74. GrayMadison T, WoulfeJohn M. Striatal blood-brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35(5):747–50.

    Article  CAS  Google Scholar 

  75. Rudolph H, Klopstein A, Gruber I, Blatti C, Lyck R, Engelhardt B. Postarrest stalling rather than crawling favors CD8(+) over CD4(+) T-cell migration across the blood-brain barrier under flow in vitro. Eur J Immunol. 2016;46(9):2187–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mosley RL, Gendelman HE, Reynolds AD, Stone DK. Proteomic studies of nitrated Alphaα-Synuclein microglia regulation by CD4+CD25+T Cells. J Proteome Res. 2009;8(7):3497–511.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Kosloski LM, Ha DM, Hutter JAL, et al. Adaptive immune regulation of glial homeostasis as an immunization strategy for neurodegenerative diseases. J Neurochem. 2010;114(5):1261–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Ma B, Xu W, Qian W, et al. Clinical correlation of peripheral CD4+-cell sub-sets, their imbalance and Parkinson's disease. Mol Med Rep. 2015;12(4 Pt.B):6105–6111.

  79. Chen Y, Qi B, Xu W, et al. Clinical correlation of peripheral CD4+-cell sub-sets, their imbalance and Parkinson’s disease. Mol Med Rep. 2015;12(4):6105–11.

    Article  CAS  PubMed  Google Scholar 

  80. Roy A, Mondal S, Kordower JH, Pahan K. Attenuation of microglial RANTES by NEMO-binding domain peptide inhibits the infiltration of CD8(+) T cells in the nigra of hemiparkinsonian monkey. Neuroscience. 2015;302:36–46.

    Article  CAS  PubMed  Google Scholar 

  81. Chandra G, Rangasamy SB, Roy A, Kordower JH, Pahan K. Neutralization of RANTES and eotaxin prevents the loss of dopaminergic neurons in a mouse model of Parkinson Disease. J Biol Chem. 2016;291(29):15267–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Nan L, Bo-Wen L, Wen-Zhi R, et al. GLP-2 Attenuates LPS-induced inflammation in BV-2 Cells by inhibiting ERK1/2, JNK1/2, and NF-κB signaling pathways. Int J Mol Sci. 2016;17(2).

  83. Guangxin C, Shoupeng F. Galangin reduces the loss of dopaminergic neurons in an LPS-evoked model of Parkinson's disease in Rats. 2017;19(1).

  84. Guangxin C. Peiminine protects dopaminergic neurons from inflammation-induced cell death by inhibiting the ERK1/2 and NF-κB signalling pathways. 2018;19(3).

  85. Sulzer D, Alcalay RN, Garretti F, et al. T cells from patients with Parkinson’s disease recognize αα-Synuclein peptides. Nature. 2017;546(7660):656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sulzer D, Alcalay RN, Garretti F, et al. Erratum: T cells from patients with Parkinson’s disease recognize αα-Synuclein peptides. Nature. 2017;549(7671):292.

    Article  CAS  PubMed  Google Scholar 

  87. Reynolds AD, Stone DK, Hutter JA, Benner EJ, Mosley RL, Gendelman HE. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson’s disease. J Immunol. 2010;184(5):2261–71.

    Article  CAS  PubMed  Google Scholar 

  88. Sommer A, Marxreiter F, Krach F, et al. th17 lymphocytes induce neuronal cell death in a human iPSC-based model of Parkinson’s disease. Cell Stem Cell. 2018;23(1):123-131.e6.

    Article  CAS  PubMed  Google Scholar 

  89. Appel SH, Beers DR, Henkel JS. T cell-microglial dialogue in Parkinson’s disease and amyotrophic lateral sclerosis: are we listening. Trends Immunol. 2010;31(1):7–17.

    Article  CAS  PubMed  Google Scholar 

  90. Harms AS, Cao S, Rowse AL, et al. MHCII is required for αα-Synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J Neurosci. 2013;33(23):9592–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. González H, Pacheco R. T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases. J Neuroinflammation. 2014;11:201.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Subbarayan MS, Hudson C, Moss LD, Nash KR, Bickford PC. T cell infiltration and upregulation of MHCII in microglia leads to accelerated neuronal loss in an α-Synuclein rat model of Parkinson’s disease. J Neuroinflammation. 2020;17(1):242.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Ferreira SA, Romero-Ramos M. Microglia response during Parkinson’s disease: alpha-Synuclein intervention. Front Cell Neurosci. 2018;12:247.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Estrada JA, Quijano-Juárez U, Contreras I. Expression of MHC-I and II by microglia and lymphocytes in the brain of diet-restricted mice. J Nutr Sci Vitaminol (Tokyo). 2019;65(2):132–41.

    Article  CAS  PubMed  Google Scholar 

  95. Milillo MA, Trotta A, Serafino A, et al. Bacterial RNA contributes to the down-modulation of MHC-II expression on monocytes/macrophages diminishing CD4+ T cell responses. Front Immunol. 2019;10:2181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Pierce S, Coetzee GA. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS ONE. 2017;12(4): e0175882.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Levite M. Dopamine and T cells: dopamine receptors and potent effects on T cells, dopamine production in T cells, and abnormalities in the dopaminergic system in T cells in autoimmune, neurological and psychiatric diseases. Acta Physiol (Oxf). 2016;216(1):42–89.

    Article  CAS  PubMed  Google Scholar 

  98. Mouzaki A, Rodi M, Dimisianos N, et al. Immune parameters that distinguish multiple sclerosis patients from patients with other neurological disorders at presentation. PLoS ONE. 2015;10(8): e0135434.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82(5):1083–94.

    Article  CAS  PubMed  Google Scholar 

  100. Rockenstein E, Ostroff G, Dikengil F, et al. Combined active humoral and cellular immunization approaches for the treatment of synucleinopathies. J Neurosci. 2018;38(4):1000–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Samantaray S, Knaryan VH, Shields DC, Cox AA, Haque A, Banik NL. Inhibition of calpain activation protects MPTP-induced nigral and spinal cord neurodegeneration, reduces inflammation, and improves Gait dynamics in mice. Mol Neurobiol. 2015;52(2):1054–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Solleiro-Villavicencio H, Rivas-Arancibia S. Effect of chronic oxidative stress on neuroinflammatory response mediated by CD4+T cells in neurodegenerative diseases. Front Cell Neurosci. 2018;12:114.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Miranda HV, Xiang W, DeOliveira RM, et al. Heat-mediated enrichment of αα-Synuclein from cells and tissue for assessing post-translational modifications. J Neurochem. 2013;126(5/6):673–84.

    Article  CAS  Google Scholar 

  104. Jęśko H, Lenkiewicz AM, Adamczyk A. Treatments and compositions targeting αα-Synuclein: a patent review (2010–2016). Expert Opin Ther Pat. 2017;27(4):427–38.

    Article  PubMed  Google Scholar 

  105. Robinson PA, Robinson PA. Understanding the molecular basis of Parkinson’s disease, identification of biomarkers and routes to therapy. Expert Rev Proteom. 2010;7(4):565–78.

    Article  CAS  Google Scholar 

  106. Moriarty GM, Janowska MK, Kang L, Baum J. Exploring the accessible conformations of N-terminal acetylated α-synuclein. FEBS Lett. 2013;587(8):1128–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Mandler M, Valera E, Rockenstein E, et al. Next-generation active immunization approach for synucleinopathies: implications for Parkinson’s disease clinical trials. Acta Neuropathol. 2014;127(6):861–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Torok N, Majlath Z, Szalardy L, Vecsei L. Investigational alphaα-Synuclein aggregation inhibitors: hope for Parkinson’s disease. Expert Opin Investig Drugs. 2016;25(11):1281–94.

    Article  CAS  PubMed  Google Scholar 

  109. Arora M, Lakshmi R. Vaccines—safety in pregnancy. Best Pract Res Clin Obstet Gynaecol. 2021;76:23–40.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Michal B, Michal B, Agmon-Levin N, Agmon-Levin N, Yehuda S, Yehuda S. Parkinson’s disease, autoimmunity and Olfaction. Int J Neurosci. 2009;119(12):2133–43.

    Article  Google Scholar 

  111. Masliah E, Valera E. Immunotherapy for neurodegenerative diseases: focus on α- synucleinopathies. Pharmacol Ther. 2013;138(3):311–22.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Jensen PH, Sanchez-Guajardo V, Annibali A, Romero-Ramos M. αα-Synuclein vaccination prevents the accumulation of Parkinson disease-like pathologic inclusions in striatum in association with regulatory T cell recruitment in a rat model. Journal of Neuropathology and Experimental Neurology 2013;72(7):624–645.

  113. Romero-Ramos M, Romero-Ramos M, Chelpin MvE, Chelpin MvE, Sanchez-Guajardo V, Sanchez-Guajardo V. Vaccination strategies for Parkinson disease. Hum Vaccin Immunother. 2014;10(4):852–867.

  114. Christiansen JR, Olesen MN, Otzen DE, Romero-Ramos M, Sanchez-Guajardo V. αα-Synuclein vaccination modulates regulatory T cell activation and microglia in the absence of brain pathology. J Neuroinflammation. 2016;13(1):74.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Lindström V, Ihse E, Fagerqvist T, et al. Immunotherapy targeting αα-Synuclein, with relevance for future treatment of Parkinson’s disease and other Lewy body disorders. Immunotherapy. 2014;6(2):141–53.

    Article  PubMed  Google Scholar 

  116. Ugen KE, Lin X, Bai G, et al. Evaluation of an α synuclein sensitized dendritic cell-based vaccine in a transgenic mouse model of Parkinson disease. Hum Vaccin Immunother. 2015;11(4):922–30.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Chen Z, Yang Y, Yang X, et al. Immune effects of optimized DNA vaccine and protective effects in a MPTP model of Parkinson’s disease. Neurol Sci. 2013;34(9):1559–70.

    Article  CAS  PubMed  Google Scholar 

  118. Gao G, Wang Z, Lu L, Duan C, Wang X, Yang H. Morphological analysis of mitochondria for evaluating the toxicity of αα-Synuclein in transgenic mice and isolated preparations by atomic force microscopy. Biomed Pharmacother. 2017;96:1380–8.

    Article  CAS  PubMed  Google Scholar 

  119. Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J. Parkinson disease and the immune system—associations, mechanisms and therapeutics. Nat Rev Neurol. 2020;16(6):303–18.

    Article  PubMed  Google Scholar 

  120. Bélanger K, Iqbal U, Tanha J, MacKenzie R, Moreno M, Stanimirovic D. Single-domain antibodies as therapeutic and imaging agents for the treatment of CNS diseases. Antibodies (Basel). 2019;8(2).

  121. Spencer B, Emadi S, Desplats P, et al. ESCRT-mediated uptake and degradation of brain-targeted αα-Synuclein single chain antibody attenuates neuronal degeneration in vivo. Mol Ther. 2014;22(10):1753–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Sardi SP, Cedarbaum JM, Brundin P. Targeted therapies for Parkinson’s disease: from genetics to the clinic. Mov Disord. 2018;33(5):684–96.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Schofield DJ, Irving L, Calo L, et al. Preclinical development of a high affinity αα-Synuclein antibody, MEDI1341, that can enter the brain, sequester extracellular αα-Synuclein and attenuate αα-Synuclein spreading in vivo. Neurobiol Dis. 2019;132: 104582.

    Article  CAS  PubMed  Google Scholar 

  124. Schenk DB, Koller M, Ness DK, et al. First-in-human assessment of PRX002, an anti-αα-Synuclein monoclonal antibody, in healthy volunteers. Mov Disord. 2017;32(2):211–8.

    Article  CAS  PubMed  Google Scholar 

  125. Jankovic J, Goodman I, Safirstein B, et al. Safety and tolerability of multiple ascending doses of PRX002/RG7935, an anti-αα-Synuclein monoclonal antibody, in patients with Parkinson disease: a randomized clinical trial. JAMA Neurol. 2018;75(10):1206–14.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Zhang W, Cao Y, Dong P, et al. Comparison of immunological characteristics of mesenchymal stem cells derived from human embryonic stem cells and bone marrow. Tissue Eng Part A. 2015;21(3/4):616–26.

    PubMed  PubMed Central  Google Scholar 

  127. Ge L, Jiang M, Duan D, et al. Secretome of olfactory mucosa mesenchymal stem cell, a multiple potential stem cell. Stem Cells Int. 2016;2016:1243659.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Zhuo Y, Wang L, Ge L, et al. Hypoxic culture promotes dopaminergic-neuronal differentiation of nasal olfactory mucosa mesenchymal stem cells via upregulation of hypoxia-inducible factor-1α. Cell Transplant. 2017;26(8):1452–61.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Hawkins KE, Corcelli M, Dowding K, et al. Embryonic stem cell-derived mesenchymal stem cells (MSCs) have a superior neuroprotective capacity over fetal MSCs in the hypoxic-ischemic mouse brain. Stem Cells Transl Med. 2018;7(5):439–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Zhou Q, Yang C, Yang P. The promotional effect of mesenchymal stem cell homing on bone tissue regeneration. Curr Stem Cell Res Ther. 2017;12(5):365–76.

    Article  CAS  PubMed  Google Scholar 

  131. Sumi K, Abe T, Kunimatsu R, et al. The effect of mesenchymal stem cells on chemotaxis of osteoclast precursor cells. J Oral Sci. 2018;60(2):221–5.

    Article  CAS  PubMed  Google Scholar 

  132. Xu W, Xu R, Li Z, Wang Y, Hu R. Hypoxia changes chemotaxis behaviour of mesenchymal stem cells via HIF-1α signalling. J Cell Mol Med. 2019;23(3):1899–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Chen L, Qiu R, Xu Q. Mesenchymal stem cell therapy for neurodegenerative diseases. J Nanosci Nanotechnol. 2014;14(1):969–75.

    Article  CAS  PubMed  Google Scholar 

  134. Offen D, Volkman R. Concise review: mesenchymal stem cells in neurodegenerative diseases. Stem Cells. 2017;35(8):1867–80.

    Article  PubMed  Google Scholar 

  135. Furno DL, Mannino G, Giuffrida R. Functional role of mesenchymal stem cells in the treatment of chronic neurodegenerative diseases. J Cell Physiol. 2018;233(5):3982–99.

    Article  PubMed  Google Scholar 

  136. Mendes Filho D, de Assuncao TSF, Oliveira LF, et al. Therapy with mesenchymal stem cells in Parkinson disease: history and perspectives. Neurologist. 2018;23(4):141–7.

    Article  PubMed  Google Scholar 

  137. Taguchi T, Ikuno M, Yamakado H, Takahashi R. Animal model for prodromal Parkinson’s disease. Int J Mol Sci. 2020;21(6).

  138. Ikuno M, Yamakado H, Akiyama H, et al. GBA haploinsufficiency accelerates alphaα-Synuclein pathology with altered lipid metabolism in a prodromal model of Parkinson’s disease. Hum Mol Genet. 2019;28(11):1894–904.

    Article  CAS  PubMed  Google Scholar 

  139. Luk KC, Kehm V, Carroll J, et al. Pathological αα-Synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338(6109):949–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med. 2008;14(5):501–3.

    Article  CAS  PubMed  Google Scholar 

  141. Xu X, Li Z, Zuo H, Chen H, Gui Y. Mesenchymal stem cell-derived exosomes altered neuron cholesterol metabolism via Wnt5a-LRP1 axis and alleviated cognitive impairment in a progressive Parkinson’s disease model. Neurosci Lett. 2022;787: 136810.

    Article  CAS  PubMed  Google Scholar 

  142. Ajmone-Cat MA, Mancini M, Simone R, Cilli P, Minghetti L. Microglial polarization and plasticity: evidence from organotypic hippocampal slice cultures. Glia. 2013;61(10):1698–711.

    Article  PubMed  Google Scholar 

  143. Single S, Merkel L, Prajeeth CK, et al. Polarized microglia do not influence oligodendrocyte lineage cells via astrocytes. Int J Dev Neurosci. 2019;77:39–47.

    Article  Google Scholar 

  144. Hoffmann A, Ettle B, Bruno A, et al. Alphaα-Synuclein activates BV2 microglia dependent on its aggregation state. Biochem Biophys Res Commun. 2016;479(4):881–6.

    Article  CAS  PubMed  Google Scholar 

  145. Song N, Wang J, Jiang H, Xie J. Astroglial and microglial contributions to iron metabolism disturbance in Parkinson's disease. Biochimica et Biophysica Acta 2018;1864(3):967–73.

  146. Park HJ, Oh SH, Kim HN, Jung YJ, Lee PH. Mesenchymal stem cells enhance alphaα-Synuclein clearance via M2 microglia polarization in experimental and human parkinsonian disorder. Acta Neuropathol. 2016;132(5):685–701.

    Article  CAS  PubMed  Google Scholar 

  147. Grant GD, Dickson TC, Pountney DL, Valdinocci D. Epothilone D inhibits microglia-mediated spread of alphaα-Synuclein aggregates. Mol Cell Neurosci. 2018;89:80–94.

    Article  PubMed  Google Scholar 

  148. Liu Y, Zhang R, Yan K, et al. Mesenchymal stem cells inhibit lipopolysaccharide-induced inflammatory responses of BV2 microglial cells through TSG-6. J Neuroinflammation. 2014;11:135.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Nash Y, Schmukler E, Trudler D, Pinkas-Kramarski R, Frenkel D. DJ-1 deficiency impairs autophagy and reduces alpha-synuclein phagocytosis by microglia. J Neurochem. 2017;143(5):584–94.

    Article  CAS  PubMed  Google Scholar 

  150. Li D, Shi J, Mao C, et al. Alteration of dynein function affects αα-synuclein degradation via the autophagosome-lysosome pathway. Int J Mol Sci. 2013;14(12):24242.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Abbott SK, Sidransky E, Murphy KE, et al. Reduced glucocerebrosidase is associated with increased αα-Synuclein in sporadic Parkinson's disease. Brain 2014;137(3): 834–48.

  152. Shen J, Wagner LK, Tong Y, Yuan Y, Huang G, Giaime E. Age-dependent dopaminergic neurodegeneration and impairment of the autophagy-lysosomal pathway in LRRK-deficient mice. Neuron. 2017;96(4):796-807.e6.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Korecka JA, Thomas R, Christensen DP, et al. Mitochondrial clearance and maturation of autophagosomes are compromised in LRRK2 G2019S familial Parkinson’s disease patient fibroblasts. Hum Mol Genet. 2019;28(19):3232–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Park HJ, Shin JY, Kim HN, Oh SH, Lee PH. Neuroprotective effects of mesenchymal stem cells through autophagy modulation in a parkinsonian model. Neurobiol Aging Exp Clin Res. 2014;35(8):1920–8.

    Article  CAS  Google Scholar 

  155. Oh SH, Lee SC, Kim DY, et al. Mesenchymal stem cells stabilize axonal transports for autophagic clearance of αα-Synuclein in Parkinsonian models. Stem Cells. 2017;35(8):1934–47.

    Article  CAS  PubMed  Google Scholar 

  156. Tolnay M, Frank S. Propagating sticky matters: An update on “prion-Like” templated misfolding in neurodegenerative disorders. Brain Pathol. 2013;23(3):319–20.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Herva ME, Spillantini MG. Parkinson’s disease as a member of Prion-like disorders. Virus Res. 2015;207:38–46.

    Article  CAS  PubMed  Google Scholar 

  158. Melki R, Bousset L, Peelaerts W, Baekelandt V. ɑα-Synuclein strains and seeding in Parkinson’s disease, incidental Lewy body disease, dementia with Lewy bodies and multiple system atrophy: similarities and differences. Cell Tissue Res. 2018;373(1):195–212.

    Article  PubMed  Google Scholar 

  159. Jiang Y, Zhang Y, Zhang L, Wang M, Zhang X, Li X. Therapeutic effect of bone marrow mesenchymal stem cells on laser-induced retinal injury in mice. Int J Mol Sci. 2014;15(6):9372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Huang Y, Tan F, Zhuo Y, et al. Hypoxia-preconditioned olfactory mucosa mesenchymal stem cells abolish cerebral ischemia/reperfusion-induced pyroptosis and apoptotic death of microglial cells by activating HIF-1α. Aging (Albany NY). 2020;12(11):10931–50.

    Article  CAS  PubMed  Google Scholar 

  161. Bi Y, Lin X, Liang H, et al. Human adipose tissue-derived mesenchymal stem cells in Parkinson’s disease: inhibition of T helper 17 cell differentiation and regulation of immune balance towards a regulatory T cell phenotype. Clin Interv Aging. 2020;15:1383–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Datta I, Bhonde R. Can mesenchymal stem cells reduce vulnerability of dopaminergic neurons in the substantia nigra to oxidative insult in individuals at risk to Parkinson’s disease. Cell Biol Int. 2012;36(7):617–24.

    Article  CAS  PubMed  Google Scholar 

  163. Zangi L, Margalit R, Reich-Zeliger S, et al. Direct imaging of immune rejection and memory induction by allogeneic mesenchymal stromal cells. Stem Cells. 2009;27(11):2865–74.

    Article  CAS  PubMed  Google Scholar 

  164. Gu LH, Zhang TT, Li Y, Yan HJ, Qi H, Li FR. Immunogenicity of allogeneic mesenchymal stem cells transplanted via different routes in diabetic rats. Cell Mol Immunol. 2015;12(4):444–55.

    Article  CAS  PubMed  Google Scholar 

  165. Huang XP, Sun Z, Miyagi Y, et al. Differentiation of allogeneic mesenchymal stem cells induces immunogenicity and limits their long-term benefits for myocardial repair. Circulation. 2010;122(23):2419–29.

    Article  CAS  PubMed  Google Scholar 

  166. Delorme B, Nivet E, Gaillard J, et al. The human nose harbors a niche of olfactory ectomesenchymal stem cells displaying neurogenic and osteogenic properties. Stem Cells Dev. 2010;19(6):853–66.

    Article  CAS  PubMed  Google Scholar 

  167. Bas E, Van De Water TR, Lumbreras V, et al. Adult human nasal mesenchymal-like stem cells restore cochlear spiral ganglion neurons after experimental lesion. Stem Cells Dev. 2014. 23(5):502–14.

  168. Hwang SH, Park SH, Choi J, et al. Age-related characteristics of multipotent human nasal inferior turbinate-derived mesenchymal stem cells. PLoS ONE. 2013;8(9): e74330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Shafiee A, Kabiri M, Ahmadbeigi N, et al. Nasal septum-derived multipotent progenitors: a potent source for stem cell-based regenerative medicine. Stem Cells Dev. 2011;20(12):2077–91.

    Article  CAS  PubMed  Google Scholar 

  170. Ge L, Zhuo Y, Duan D, et al. Biological characteristics of human olfactory mucosa mesenchymal stem cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2015;40(1):53–8.

    CAS  PubMed  Google Scholar 

  171. Moya A, Paquet J, Deschepper M, et al. Human mesenchymal stem cell failure to adapt to glucose shortage and rapidly use intracellular energy reserves through glycolysis explains poor cell survival after implantation. Stem Cells. 2018;36(3):363–76.

    Article  CAS  PubMed  Google Scholar 

  172. Hu C, Li L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J Cell Mol Med. 2018;22(3):1428–42.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med. 2019;23(2):720–30.

    Article  PubMed  Google Scholar 

  174. Li L, Wu S, Li P, Zhuo L, Gao Y, Xu Y. Hypoxic preconditioning combined with microbubble-mediated ultrasound effect on MSCs promote SDF-1/CXCR4 expression and its migration ability: an in vitro study. Cell Biochem Biophys. 2015;73(3):749–57.

    Article  PubMed  Google Scholar 

  175. Meng SS, Xu XP, Chang W, et al. LincRNA-p21 promotes mesenchymal stem cell migration capacity and survival through hypoxic preconditioning. Stem Cell Res Ther. 2018;9(1):280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Redondo-Castro E, Cunningham C, Miller J, et al. Interleukin-1 primes human mesenchymal stem cells towards an anti-inflammatory and pro-trophic phenotype in vitro. Stem Cell Res Ther. 2017;8(1):79.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Ren G, Zhao X, Zhang L, et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol. 2010;184(5):2321–8.

    Article  CAS  PubMed  Google Scholar 

  178. Liu D, Kong F, Yuan Y, et al. Decorin-modified umbilical cord mesenchymal stem cells (Mscs) attenuate radiation-induced lung injuries via regulating inflammation, fibrotic factors, and immune responses. Int J Radiat Oncol Biol Phys. 2018;101(4):945–56.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Yaxuan He from UCLA for the revision of English grammar and native language. We thank International Science Editing (http://www.internationalscienceediting.com) for editing this manuscript.

Funding

This study was supported by Grants: Key Research and Development Program of Hunan Province of China (No. 2020SK2102). Supported by Changsha Municipal Natural Science Foundation (No. kq2208154). Scientific Research Project of Hunan Provincial Health Commission (No. B202304088074). Scientific Research Project of Hunan Provincial Health Commission (No. 202203105045).

Author information

Authors and Affiliations

Authors

Contributions

YZ is responsible for the conception and design and writing the initial manuscript and its editing. XL is responsible for the production and design of figures and for writing the initial manuscript. ML and ZWH are responsible for the conception and design. All authors declare a contribution to this paper. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Zhengwen He or Ming Lu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

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

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

Zhuo, Y., Li, X., He, Z. et al. Pathological mechanisms of neuroimmune response and multitarget disease-modifying therapies of mesenchymal stem cells in Parkinson’s disease. Stem Cell Res Ther 14, 80 (2023). https://doi.org/10.1186/s13287-023-03280-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-023-03280-0

Keywords