Next Article in Journal
“Build Your Village”—Conducting the Village Test on Cognitively Impaired Patients: A First Journey into Alzheimerland
Previous Article in Journal
Sex-Related Differences in Post-Stroke Anxiety, Depression and Quality of Life in a Cohort of Smokers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson’s Disease

by
Mary Ann Chapman
1,* and
Barbara A. Sorg
2
1
Visage Communications, Spokane, WA 99021, USA
2
R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2024, 14(6), 522; https://doi.org/10.3390/brainsci14060522
Submission received: 17 April 2024 / Revised: 13 May 2024 / Accepted: 15 May 2024 / Published: 21 May 2024
(This article belongs to the Section Neurodegenerative Diseases)

Abstract

:
The role of the extracellular matrix (ECM) in Parkinson’s disease (PD) is not well understood, even though it is critical for neuronal structure and signaling. This systematic review identified the top deregulated ECM-related pathways in studies that used gene set enrichment analyses (GSEA) to document transcriptomic, proteomic, or genomic alterations in PD. PubMed and Google scholar were searched for transcriptomics, proteomics, or genomics studies that employed GSEA on data from PD tissues or cells and reported ECM-related pathways among the top-10 most enriched versus controls. Twenty-seven studies were included, two of which used multiple omics analyses. Transcriptomics and proteomics studies were conducted on a variety of tissue and cell types. Of the 17 transcriptomics studies (16 data sets), 13 identified one or more adhesion pathways in the top-10 deregulated gene sets or pathways, primarily related to cell adhesion and focal adhesion. Among the 8 proteomics studies, 5 identified altered overarching ECM gene sets or pathways among the top 10. Among the 4 genomics studies, 3 identified focal adhesion pathways among the top 10. The findings summarized here suggest that ECM organization/structure and cell adhesion (particularly focal adhesion) are altered in PD and should be the focus of future studies.

1. Introduction

The extracellular matrix (ECM) is a structural and functional complex that surrounds and anchors nearly all cells in the body. The core matrisome consists of approximately 300 proteins that have been grouped broadly into collagens, glycoproteins, and proteoglycans [1]. The presence and ratio of these proteins vary by organ and tissue type [1,2]. Compared with other organs, the brain is relatively poor in fibrous proteins such as collagen but rich in proteoglycans that contain negatively charged glycosaminoglycan (GAG) side chains that sequester cations and water.
Beyond giving the brain its gelatinous texture, the ECM plays numerous functional roles. Through its binding to integrins in cell membranes and its downstream effects on the cytoskeleton, the ECM signals cell survival and maintains differentiation, polarity, and morphology [3,4]. The ECM also regulates growth factor function through binding and releasing growth factor proteins and their fragments. For example, the ECM component heparan sulfate serves as a low-affinity binding site for basic fibroblast growth factor (bFGF), which is essential for its high-affinity binding [5]. Signaling by glial cell line-derived neurotrophic factor (GDNF) requires heparan sulfate glycosaminoglycan [6], and the heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF and other GDNF family ligands that mediate cell spreading and neurite outgrowth [7].
Another key function of the ECM is in the formation and plasticity of synapses. Many recent plasticity investigations have centered on perineuronal nets, ECM structures comprising chondroitin sulfate proteoglycans, link proteins, hyaluronic acid, and tenascins that surround the selected somas and proximal dendrites [8,9]. Perineuronal nets appear during the critical period of synaptic development and help stabilize synapses, although they alter plasticity, even in adulthood [10,11]. This plasticity has been demonstrated in various neuronal systems and models through the digestion of chondroitin sulfate, which reactivates ocular dominance plasticity in adult rats [12] and restores memory in mice with tau pathology [13]. However, the function of perineuronal nets on plasticity may be more complex, as digestion of perineuronal nets can also impair learning and memory [10]. Other ECM arrangements are also found in the nervous system, including diffuse ECM containing axonal coats [14] and perinodal ECM [15], although their roles have been studied less.
As an integral component of brain tissue that dynamically interacts with neurons and glia, the ECM is expected to be affected by, and contribute to, neurodegenerative processes in diseases such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). Indeed, over the past decade, evidence has accumulated that implicates the ECM in neurodegeneration [16] and PD specifically [17,18]. Studies that find such effects have tended to analyze transcriptomics, proteomics, and genomics data using pathway approaches or gene sets instead of individual protein or gene comparisons. Pathway analyses permit the assessment of cumulative changes throughout individual biological processes that may, together, exert an effect on cell or tissue structure and function. This systematic review was, therefore, designed to identify the top deregulated ECM-related pathways in studies that used gene set enrichment analysis to document transcriptomic, proteomic, or genomic alterations in PD.

2. Materials and Methods

The PubMed database was searched for research reports using the following search strategy: Parkinson’s disease AND (proteomics OR GWAS OR gene expression OR RNA sequencing analysis OR enrichment OR pathways OR KEGG OR gene ontology OR Reactome) AND (extracellular matrix OR adhesion OR collagen OR proteoglycans OR glycosaminoglycans). Given that searches in Google Scholar are organized differently from those in PubMed, the following advanced search strategy was used in Google Scholar: Parkinson disease (all of the words), extracellular matrix (exact phrase), and at least one of the following words, where the words were listed anywhere in the article: KEGG OR ontology OR Reactome. The results were sorted by relevance. The title and/or abstract of each result was read to identify relevant studies, and the reference lists of studies and review articles were consulted for additional articles that the searches missed. This study was not registered on Prospero.
Articles were included if they (1) described published, peer-reviewed research studies of transcriptomics, proteomics, or genomics in PD, (2) were conducted using tissues or cells derived from humans with PD, (3) employed gene set enrichment analysis (GSEA), and (4) identified one or more ECM-related pathway among the top-10 enriched gene sets. Reviews, preprints, and studies that evaluated ECM in animal models of PD were excluded, although very few studies have reported changes in the ECM in animal models of PD [19,20,21], with the major focus on matrix metalloproteinases (MMPs), which regulate the ECM [22,23,24]; for review, see [25]. Studies conducted in multiple disease populations were also excluded unless results for Parkinson’s disease patients were reported separately.
Articles that met the criteria were reviewed, and study information was extracted into a summary table. Gene sets considered ECM-related were adhesion groups or pathways (adherens junctions, cell adhesion, cell adhesion molecules, cell adhesion substrate, and focal adhesions) and those that had ECM, collagen, integrin, GAG, or proteoglycan in the pathway name (e.g., ECM organization). ECM-related gene sets or pathways reported by the authors of each study to be among the top-10 deregulated or enriched (as defined by the study authors) were recorded in the summary table.

3. Results

The PubMed search yielded 365 results, 19 of which met the inclusion criteria. The Google Scholar search yielded 8670 results; after the first 300 results in the Google Scholar search, relevance declined dramatically, and, thus, the remaining pages were not considered. Five additional articles identified in the Google Scholar search but not in the PubMed search met the criteria. Searches of the reference lists of relevant articles identified 3 additional studies with prominent ECM findings, for a total of 27 studies (Figure 1). All studies included one or more control groups.
Studies could be classified as transcriptomics, proteomics, or genomics; two studies used multiple omics analyses and are, therefore, entered into the tables twice (in different sections) (Table 1, Supplemental Table S1). The gene set enrichment analyses were conducted using various databases, such as Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome, and gene ontology (GO) terms. Top pathways were identified by the authors of each article based on p values adjusted for multiple testing or q values.

3.1. Transcriptomics Studies

The 17 transcriptomics studies (16 data sets) identified in the search were conducted on a variety of human cell and tissue types and included both monogenic and idiopathic PD (Table 1 and Supplemental Table S1) [17,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. Most of the studies performed gene set enrichment analysis using GO terms or KEGG pathways, but three used Reactome, Metabase Ontology, or Pathway Studio (Figure 2; Supplemental Table S1). The studies assessed deregulated pathways in induced pluripotent stem cells (iPSCs) [26,27,28,32], dermal fibroblasts [17,33], mesenchymal stem cells [30], blood [29,31,34,35], and various brain regions [36,37,38,39,40,41].
Thirteen of the sixteen data sets identified one or more adhesion pathways in the top-10 deregulated gene sets or pathways (Figure 2). These were primarily related to cell adhesion (8 of 16 data sets) and focal adhesion/integrin binding (5 of 16 data sets). Four studies identified altered collagen gene sets or pathways, and nine identified overarching ECM gene sets or pathways among the top 10. One study reported somewhat different results for iPSC cell lines from those with sporadic PD versus PD associated with GBA1 mutations [26].

3.2. Proteomics Studies

The eight proteomics studies identified in the search were also conducted on a variety of human cell and tissue types and included monogenic and idiopathic PD (Table 1 and Supplemental Table S1) [18,36,42,43,44,45,46,47]. These studies identified deregulated collagen proteins or collagen pathways in iPSCs [42], cerebrospinal fluid [45] tears [44], plasma [46], serum exosomes [47], and various brain regions [18,36,43].
Only two of the proteomics studies identified an adhesion pathway among the top-10 deregulated pathways [36,43], although another found three integrin signaling pathways among the top five [46], which are related to focal adhesions (Figure 3). Five of the eight proteomics studies identified altered overarching ECM gene sets or pathways among the top 10, and three of the six identified GAG or proteoglycan pathways among the top 10 (Figure 3).

3.3. Genomics Studies

Four genomics studies were identified in the search, which included patients with familial and idiopathic PD (Table 1, Supplemental Table S1) [39,48,49,50]. Three of these four studies identified adhesion pathways among the top 10 most enriched, and all of them included the focal adhesion pathway (Figure 4). In the fourth study, ECM constituents and ECM disassembly were the only gene sets in the entire study identified as statistically significant [48]. The genomics studies did not identify any GAG- or proteoglycan-related gene sets among the top 10.

4. Discussion

This review documents the frequent alterations in ECM-related pathways reported in transcriptomics, proteomics, and genomics studies of PD. The transcriptomics and proteomics studies were conducted with different cell and tissue types, and in different brain regions, in accord with the involvement of multiple tissues and neural systems in PD and the plethora of motor and non-motor symptoms [51,52]. The studies also spanned different PD populations, including those with sporadic PD and variations in PD-related genes, such as LRRK-2, Parkin, and SNCA.
Among the different types of omics studies, ECM findings in GWAS pathway studies were the most consistent and were documented in several demographically disparate populations [39,49]. The focal adhesion pathway—dynamic protein complexes that connect the cytoskeleton to the ECM—was among the top-10 pathways in three of the four genomics studies reviewed here. These findings are supported by a recent, novel modeling approach, designed to explore disease mechanisms based on genomic data, which also identified focal adhesion as a top enriched pathway in PD [53]. Alterations in individual ECM genes have rarely been reported in PD, although scattered exceptions exist, such as the GPNMB gene [54,55,56], which is involved in cell adhesion [57]. This suggests that numerous, less frequent variations across multiple ECM genes drive the significance in the GWAS pathway analyses and may collectively constitute a risk factor for PD.
Adhesion gene sets and pathways were also among the top pathways identified in the transcriptomics studies (13 of 16 data sets) in this review. These findings are supported by those of Stern and colleagues, who have consistently reported deregulation of the focal adhesion pathway in their studies with iPSC-derived neurons [26,28,58,59], as well as in a recent systematic review of deregulated proteins in PD brains [60]. Here, we extend their findings to include studies that reported alterations in adhesion gene sets or pathways beyond the brain. For example, cell or focal adhesion alterations were noted in studies on iPSC-derived dopamine neurons and astrocytes, mesenchymal stem cells, dermal fibroblasts, whole blood, and brain. These observations suggest that ECM alterations are not necessarily restricted to areas of cell loss in the PD brain but are likely a more general disease-related phenomenon. The observation of adhesion deregulation in fibroblasts [17] is particularly interesting as fibroblasts show increased growth in PD [61] rather than degeneration.
Transcriptomic and proteomic studies included in the present review also frequently identified alterations in collagen and ECM structural and organizational gene sets/pathways. These alterations are typified by the description in the gene ontology set for ECM organization: “a process that is carried out at the cellular level which results in the assembly, arrangement of constituent parts, or disassembly of an extracellular matrix [62]”. These gene sets and pathways include proteins, such as collagens, transforming growth factor-beta (TGF-beta), matrix metalloproteinases (MMPs), and disintegrin and metalloproteases (ADAMs), as well proteins that form the basement membrane lining epithelial and endothelial tissues, such as the blood vessels. Of the few studies that have specifically assessed the ECM in PD, alterations in MMPs and basement membrane proteins are among the most frequent findings [63,64,65,66,67].
Few studies in the present review identified alterations in glycosaminoglycan (GAG) or proteoglycan gene sets/pathways. These ECM components play critical structural roles in cells and regulate cell signaling by binding and releasing growth factors [1]. However, GAGs and proteoglycans are difficult to study because of their extensive glycosylation, which is critical to their function but difficult to measure, particularly using proteomics techniques [64,65]. Novel methods may be needed to adequately explore the role of these biochemicals in PD and other diseases [64]; some of these glycomic and glycoproteomic methods are described in a recent review [68].
The results described here add to a growing literature documenting deregulation of ECM-related pathways in PD. The findings in genomics, transcriptomics, and proteomics studies conducted in non-neuronal tissues suggest that ECM-related alterations may not be a response to PD medications, which has been a concern with brain tissue. Moreover, ECM-related alterations have been identified in several studies investigating the role of alpha-synuclein [69,70], a protein frequently misfolded and aggregated in PD brains [71] and the genetic variations of which are causes and/or risk factors for PD [72]. These studies found that ECM-related pathways were among the top-5 deregulated pathways in human cell lines that overexpressed alpha synuclein [69,70]. Additional interactions among ECM molecules and alpha synuclein have been documented in animal and in vitro studies [73,74,75,76,77,78,79,80]. For instance, in a study of oligodendrocyte progenitor cells, overexpression of alpha synuclein led to integrin down-regulation that reduced adhesion to fibronectin and led to cell death [81]. Additionally, a GSEA analysis of a human cell line found that alpha synuclein overexpression led to reduced expression of ECM, collagen, and heparin gene sets [70].
Several lines of research have focused on the interactions between the ECM and extracellular alpha synuclein based on evidence that alpha synuclein can be secreted from cells via exosomes in an activity-dependent manner [82,83]. One area of investigation is the identification of mechanisms by which alpha synuclein activates microglia, initiating a proinflammatory response characterized by elevated cytokine production [84,85]. At least one of the receptors involved in microglial activation is the integrin CD11b (the alpha chain of integrin αMβ2); alpha synuclein binding to integrin CD11b activates microglial NADPH oxidase (NOX2) through a RhoA-dependent pathway, a potential mechanism by which extracellular alpha synuclein could cause neuronal damage and death [86].
Other research has focused on interactions between extracellular alpha synuclein and GAGs or proteoglycans as a potential mechanism of prion-like spreading and seeding of alpha synuclein aggregates [18], which has been hypothesized to underlie PD progression [87]. GAGs are localized to Lewy bodies in PD [88] and inhibit proteases that degrade alpha synuclein [89]. Moreover, the internalization of alpha synuclein fibrils into neuroblastoma and oligodendrocyte-like cells depends on heparan sulfate [76]. Other findings emphasize the potential protective nature of heparin in synuclein pathology. For instance, using electron microscopy, a recent in vitro study visualized alpha synuclein fibrils complexed with heparin, the presence of which altered fibril morphology and led to reduced pathological aggregation of endogenous alpha synuclein in primary cortical neurons [90].
In addition to direct interactions between the ECM and extracellular alpha synuclein, functional intracellular interactions likely occur at the synapse via the cytoskeleton. Synaptic dysfunction is an early pathological event in PD, marked by decreased striatal dopamine synthesis, storage, release, and transporter binding [91]. Wild-type alpha synuclein is concentrated at synapses, where it binds actin, slowing polymerization and accelerating depolymerization; in contrast, the PD-associated A30P mutant alpha synuclein increases actin polymerization and disrupts the cytoskeleton during the reassembly of actin filaments [92]. The ECM is a major regulator of the cytoskeletal and synaptic structure via integrin binding [93], and the focal adhesion pathway is among the most frequently dysregulated in PD [60]. Based on these observations, investigations into the interactions between ECM/focal adhesions and alpha synuclein at the synaptic cytoskeleton appear warranted.
Notably, ECM-related alterations are not specific to PD, as evidence from published omics studies that examined multiple neurodegenerative diseases has also documented the significance of ECM-related pathways, including adhesion pathways [94,95]. An analysis of 16 proteomics studies of PD, AD, and Huntington’s disease (HD) based on postmortem brain tissue found dysregulation of various biological processes across the three diseases, including ECM assembly and organization, basement membrane organization, and metabolism of GAGs and proteoglycans [65]. The only individual protein altered across all three diseases was collagen type I alpha 2, a gene expressed primarily in connective tissue but also found in the brain, where it drives glioblastoma progression [96]. Another bioinformatics study analyzed a database of differentially expressed miRNAs in body fluids and circulating cells of patients with PD, AD, amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) from 72 studies. They found that pathways consistently targeted by several miRNAs included ECM receptor interactions, adherens junctions, and TGF-beta signaling [97]. The regulation of cell matrix adhesion was among the top protein–protein interaction pathways. An analysis of transcriptomic data from AD, PD, HD, and ALS studies found that ECM organization was among the top-8 overrepresented biological processes across the diseases, and all six detected cellular component GSEA terms related to focal adhesion, plasma membrane, and endoplasmic reticulum lumen [94]. These findings suggest that ECM-related pathways are among those broadly affected in neurodegenerative disease.
Given that dopaminergic neurons in the substantia nigra, pars compacta (SNpc) are much more susceptible to death in PD than those in the ventral tegmental area (VTA) [98], researchers have sought to identify differences in gene or protein expression that may protect VTA neurons or predispose SNpc neurons. A recent study by Aguila et al. [99] examined transcriptomics in VTA and SNpc dopamine neurons in 18 human postmortem brains from individuals without neurological disease. In their study, the most highly enriched pathways in the VTA included ECM modulation, cytoskeletal regulation, and synapse formation, among several others. These related pathways were frequently among the top deregulated sets in the studies reviewed here and suggest that coordinated alterations in synaptic morphology and structure may be more damaging in the SNpc than in the VTA.
The GSEA findings described in this review exemplify the utility of omics studies, which provide an unbiased analysis of genetic variation and protein dysregulation, pointing to understudied pathways in disease. ECM and related pathways such as focal and cell adhesion are essential for neuronal development and structure, synaptic function, and cell survival [100,101,102,103]. However, the roles of ECM-related pathways and components remain understudied in PD and possibly other neurodegenerative diseases. In PD, synaptic dysfunction is an early pathological event that precedes neurodegeneration [91,104,105,106]. Given that ECM-related pathways are critical for synaptic function, stability, and plasticity [107], they may be targets for early intervention in PD.
The question then becomes which molecules to target, given that the genomic, transcriptomic, and proteomic alterations are present throughout ECM-related pathways. The answer to this question will be informed by further research into specific molecules and pathways and may involve cocktails that target multiple ECM-related molecules. Given their role as effectors, kinases are frequent therapeutic targets and have been pursued in PD [108]. Indeed, mutations in leucine repeat rich kinase-2 (LRRK2) are common risk factors for PD, and an LRRK2 inhibitor is in late-stage PD clinical trials [109]. However, kinases related more directly to the ECM and cell adhesion such as focal adhesion kinase have not been explored and may be potential targets if selectivity for affected brain regions could be achieved. Individual ECM-related molecules that may be potential therapeutic targets are suggested by various studies described earlier, including heparin and CD11b integrin. The intersection of ECM, cytoskeleton, synapse integrity, and synaptic vesicle recycling appears repeatedly in PD studies [110] and differentiates the VTA and SN [99], implying that cross-pathway research into synaptic dysfunction may provide clues to PD etiopathology and, ultimately, treatment. A related line of research involves the detachment of neurons from the ECM, which leads to cell death via anoikis—a type of programmed cell death. A recent diagnostic model of PD has been developed based on the differential expression of anoikis-related genes in human blood, implicating detachment from the ECM as an important factor in PD and suggesting an intervention point [111].
The present review has several limitations that are important to note. First, in most of the studies reviewed here, many other pathways than those related to the ECM were enriched. Indeed, the interpretation of omics pathway studies can be complex given the variety and sometimes apparently unrelated pathways identified as significant. Second, this review excluded studies that did not report significant ECM-related findings, as per the search criteria. A subsequent exploratory literature search designed to identify omics studies in PD that did not identify ECM-related pathways as top hits resulted in more than 20 studies [45,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130]. However, upon closer review of these studies, it was noted that some limited the proteins or pathways that were included in the final models/reports for various reasons and, therefore, may not be expected to find ECM alterations (e.g., removal of genes expressed at low levels [112], inclusion limited to pathways with established relevance to PD [117]). Several other excluded studies reported significant findings for one or more individual ECM components rather than as part of a gene set [45,114,115] or as a significant interaction pathway rather than as part of a primary altered pathway [126]. Thus, although a substantial number of published omics studies do not report ECM-related pathways as significant, there is still ample reason to suspect their involvement and alteration.
An additional limitation of the present review relates to the small sample sizes of some of the included studies. Aguila and colleagues describe the importance of determining adequate sample sizes and using large-enough cohorts to reliably identify relevant DEGs; for their transcriptomic analysis, the minimum sample size needed was N = 8 [99]. This review did not take into consideration sample size, which could have led to spurious findings in some of the studies. Thus, caution is warranted in interpreting results from studies based on small sample sizes [99]. A related caution is the implicit assumption that gene sets/pathways with the lowest p values are the most important to the cell. Ranking gene sets according to p values does not necessarily correspond to the magnitude of their effects in the cell or their contribution to disease. These limitations underscore the need to complement findings from omics and bioinformatics analyses with in vitro and in vivo research. Based on the omics studies reviewed here, ECM-related pathways seem to be excellent candidates for such work.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/brainsci14060522/s1. Table S1: Data extracted from studies included in this article.

Author Contributions

Conceptualization, M.A.C. and B.A.S.; methodology, M.A.C.; writing—original draft preparation, M.A.C.; writing—review and editing, B.A.S.; visualization, M.A.C.; funding acquisition, B.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by the National Institutes of Health DA 055645 and the Good Samaritan Foundation of Legacy Health.

Data Availability Statement

The data supporting the findings of this article are available within the article and its Supplementary Material.

Conflicts of Interest

Mary Ann Chapman owns and is employed by Visage Communications. Over the past 5 years, she has received support from AbbVie, the Alliance for Patient Access, the Clinical Neurological Society of America, TTC Group, and the International Association of Parkinsonism and Related Disorders for work unrelated to the work under consideration. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Hynes, R.O.; Naba, A. Overview of the matrisome—An inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012, 4, a004903. [Google Scholar] [CrossRef] [PubMed]
  2. Karamanos, N.K.; Theocharis, A.D.; Piperigkou, Z.; Manou, D.; Passi, A.; Skandalis, S.S.; Vynios, D.H.; Orian-Rousseau, V.; Ricard-Blum, S.; Schmelzer, C.E.H.; et al. A guide to the composition and functions of the extracellular matrix. FEBS J. 2021, 288, 6850–6912. [Google Scholar] [CrossRef] [PubMed]
  3. Meredith, J.E., Jr.; Fazeli, B.; Schwartz, M.A. The extracellular matrix as a cell survival factor. Mol. Biol. Cell 1993, 4, 953–961. [Google Scholar] [CrossRef]
  4. Kim, S.H.; Turnbull, J.; Guimond, S. Extracellular matrix and cell signalling: The dynamic cooperation of integrin, proteoglycan and growth factor receptor. J. Endocrinol. 2011, 209, 139–151. [Google Scholar] [CrossRef] [PubMed]
  5. Yayon, A.; Klagsbrun, M.; Esko, J.D.; Leder, P.; Ornitz, D.M. Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor. Cell 1991, 64, 841–848. [Google Scholar] [CrossRef] [PubMed]
  6. Barnett, M.W.; Fisher, C.E.; Perona-Wright, G.; Davies, J.A. Signalling by glial cell line-derived neurotrophic factor (GDNF) requires heparan sulphate glycosaminoglycan. J. Cell Sci. 2002, 115, 4495–4503. [Google Scholar] [CrossRef] [PubMed]
  7. Bespalov, M.M.; Sidorova, Y.A.; Tumova, S.; Ahonen-Bishopp, A.; Magalhães, A.C.; Kulesskiy, E.; Paveliev, M.; Rivera, C.; Rauvala, H.; Saarma, M. Heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF, neurturin, and artemin. J. Cell Biol. 2011, 192, 153–169. [Google Scholar] [CrossRef] [PubMed]
  8. Sorg, B.A.; Berretta, S.; Blacktop, J.M.; Fawcett, J.W.; Kitagawa, H.; Kwok, J.C.; Miquel, M. Casting a Wide Net: Role of Perineuronal Nets in Neural Plasticity. J. Neurosci. 2016, 36, 11459–11468. [Google Scholar] [CrossRef] [PubMed]
  9. Chaunsali, L.; Tewari, B.P.; Sontheimer, H. Perineuronal Net Dynamics in the Pathophysiology of Epilepsy. Epilepsy Curr. 2021, 21, 273–281. [Google Scholar] [CrossRef]
  10. Fawcett, J.W.; Fyhn, M.; Jendelova, P.; Kwok, J.C.F.; Ruzicka, J.; Sorg, B.A. The extracellular matrix and perineuronal nets in memory. Mol. Psychiatry 2022, 27, 3192–3203. [Google Scholar] [CrossRef]
  11. Fawcett, J.W.; Oohashi, T.; Pizzorusso, T. The roles of perineuronal nets and the perinodal extracellular matrix in neuronal function. Nat. Rev. Neurosci. 2019, 20, 451–465. [Google Scholar] [CrossRef] [PubMed]
  12. Pizzorusso, T.; Medini, P.; Berardi, N.; Chierzi, S.; Fawcett, J.W.; Maffei, L. Reactivation of ocular dominance plasticity in the adult visual cortex. Science 2002, 298, 1248–1251. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, S.; Cacquevel, M.; Saksida, L.M.; Bussey, T.J.; Schneider, B.L.; Aebischer, P.; Melani, R.; Pizzorusso, T.; Fawcett, J.W.; Spillantini, M.G. Perineuronal net digestion with chondroitinase restores memory in mice with tau pathology. Exp. Neurol. 2015, 265, 48–58. [Google Scholar] [CrossRef]
  14. Bruckner, G.; Morawski, M.; Arendt, T. Aggrecan-based extracellular matrix is an integral part of the human basal ganglia circuit. Neuroscience 2008, 151, 489–504. [Google Scholar] [CrossRef]
  15. Bekku, Y.; Oohashi, T. Neurocan contributes to the molecular heterogeneity of the perinodal ECM. Arch. Histol. Cytol. 2010, 73, 95–102. [Google Scholar] [CrossRef]
  16. Bonneh-Barkay, D.; Wiley, C.A. Brain extracellular matrix in neurodegeneration. Brain Pathol. 2009, 19, 573–585. [Google Scholar] [CrossRef] [PubMed]
  17. Fernández-Santiago, R.; Esteve-Codina, A.; Fernández, M.; Valldeoriola, F.; Sanchez-Gómez, A.; Muñoz, E.; Compta, Y.; Tolosa, E.; Ezquerra, M.; Martí, M.J. Transcriptome analysis in LRRK2 and idiopathic Parkinson’s disease at different glucose levels. NPJ Parkinsons Dis. 2021, 7, 109. [Google Scholar] [CrossRef] [PubMed]
  18. Raghunathan, R.; Hogan, J.D.; Labadorf, A.; Myers, R.H.; Zaia, J. A glycomics and proteomics study of aging and Parkinson’s disease in human brain. Sci. Rep. 2020, 10, 12804. [Google Scholar] [CrossRef]
  19. Jung, S.Y.; Choi, J.M.; Rousseaux, M.W.C.; Malovannaya, A.; Kim, J.J.; Kutzera, J.; Wang, Y.; Huang, Y.; Zhu, W.; Maity, S.; et al. An Anatomically Resolved Mouse Brain Proteome Reveals Parkinson Disease-relevant Pathways. Mol. Cell Proteom. 2017, 16, 581–593. [Google Scholar] [CrossRef]
  20. Villadiego, J.; García-Swinburn, R.; García-González, D.; Lebrón-Galán, R.; Murcia-Belmonte, V.; García-Roldán, E.; Suárez-Luna, N.; Nombela, C.; Marchena, M.; de Castro, F.; et al. Extracellular matrix protein anosmin-1 overexpression alters dopaminergic phenotype in the CNS and the PNS with no pathogenic consequences in a MPTP model of Parkinson’s disease. Brain Struct. Funct. 2023, 228, 907–920. [Google Scholar] [CrossRef]
  21. Khan, A.H.; Lee, L.K.; Smith, D.J. Single-cell analysis of gene expression in the substantia nigra pars compacta of a pesticide-induced mouse model of Parkinson’s disease. Transl. Neurosci. 2022, 13, 255–269. [Google Scholar] [CrossRef] [PubMed]
  22. Chung, Y.C.; Kim, Y.S.; Bok, E.; Yune, T.Y.; Maeng, S.; Jin, B.K. MMP-3 contributes to nigrostriatal dopaminergic neuronal loss, BBB damage, and neuroinflammation in an MPTP mouse model of Parkinson’s disease. Mediators Inflamm. 2013, 2013, 370526. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, Y.S.; Joh, T.H. Matrix metalloproteinases, new insights into the understanding of neurodegenerative disorders. Biomol. Ther. 2012, 20, 133–143. [Google Scholar] [CrossRef] [PubMed]
  24. Annese, V.; Herrero, M.-T.; Di Pentima, M.; Gomez, A.; Lombardi, L.; Ros, C.M.; De Pablos, V.; Fernandez-Villalba, E.; De Stefano, M.E. Metalloproteinase-9 contributes to inflammatory glia activation and nigro-striatal pathway degeneration in both mouse and monkey models of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. Brain Struct. Funct. 2015, 220, 703–727. [Google Scholar] [CrossRef] [PubMed]
  25. Behl, T.; Kaur, G.; Sehgal, A.; Bhardwaj, S.; Singh, S.; Buhas, C.; Judea-Pusta, C.; Uivarosan, D.; Munteanu, M.A.; Bungau, S. Multifaceted Role of Matrix Metalloproteinases in Neurodegenerative Diseases: Pathophysiological and Therapeutic Perspectives. Int. J. Mol. Sci. 2021, 22, 1413. [Google Scholar] [CrossRef] [PubMed]
  26. Rosh, I.; Tripathi, U.; Hussein, Y.; Rike, W.A.; Djamus, J.; Shklyar, B.; Manole, A.; Houlden, H.; Winkler, J.; Gage, F.H.; et al. Synaptic dysfunction and extracellular matrix dysregulation in dopaminergic neurons from sporadic and E326K-GBA1 Parkinson’s disease patients. NPJ Parkinsons Dis. 2024, 10, 38. [Google Scholar] [CrossRef] [PubMed]
  27. Akrioti, E.; Karamitros, T.; Gkaravelas, P.; Kouroupi, G.; Matsas, R.; Taoufik, E. Early Signs of Molecular Defects in iPSC-Derived Neural Stems Cells from Patients with Familial Parkinson’s Disease. Biomolecules 2022, 12, 876. [Google Scholar] [CrossRef] [PubMed]
  28. Stern, S.; Lau, S.; Manole, A.; Rosh, I.; Percia, M.M.; Ben Ezer, R.; Shokhirev, M.N.; Qiu, F.; Schafer, S.; Mansour, A.A.; et al. Reduced synaptic activity and dysregulated extracellular matrix pathways in midbrain neurons from Parkinson’s disease patients. NPJ Parkinsons Dis. 2022, 8, 103. [Google Scholar] [CrossRef] [PubMed]
  29. Hemmings, S.M.J.; Swart, P.; Womersely, J.S.; Ovenden, E.S.; van den Heuvel, L.L.; McGregor, N.W.; Meier, S.; Bardien, S.; Abrahams, S.; Tromp, G.; et al. RNA-seq analysis of gene expression profiles in posttraumatic stress disorder, Parkinson’s disease and schizophrenia identifies roles for common and distinct biological pathways. Discov. Ment. Health 2022, 2, 6. [Google Scholar] [CrossRef]
  30. Cho, E.; Park, J.; Kim, K.; Kim, M.G.; Cho, S.R. Reelin Alleviates Mesenchymal Stem Cell Senescence and Reduces Pathological alpha-Synuclein Expression in an In Vitro Model of Parkinson’s Disease. Genes 2021, 12, 1066. [Google Scholar] [CrossRef]
  31. Yang, M.; Wu, X.Q.; Ding, C.B.; Zhang, G.F.; Li, M.; Lv, L.N.; Li, Y.H.; Sun, D.W.; Zhao, J.J. Weighted gene co-expression network analysis identifies specific modules and hub genes related to Parkinson’s disease. Neuroreport 2021, 32, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  32. Booth, H.D.E.; Wessely, F.; Connor-Robson, N.; Rinaldi, F.; Vowles, J.; Browne, C.; Evetts, S.G.; Hu, M.T.; Cowley, S.A.; Webber, C.; et al. RNA sequencing reveals MMP2 and TGFB1 downregulation in LRRK2 G2019S Parkinson’s iPSC-derived astrocytes. Neurobiol. Dis. 2019, 129, 56–66. [Google Scholar] [CrossRef] [PubMed]
  33. González-Casacuberta, I.; Moren, C.; Juarez-Flores, D.L.; Esteve-Codina, A.; Sierra, C.; Catalan-Garcia, M.; Guitart-Mampel, M.; Tobias, E.; Milisenda, J.C.; Pont-Sunyer, C.; et al. Transcriptional alterations in skin fibroblasts from Parkinson’s disease patients with parkin mutations. Neurobiol. Aging 2018, 65, 206–216. [Google Scholar] [CrossRef] [PubMed]
  34. Tan, C.; Liu, X.; Chen, J. Microarray Analysis of the Molecular Mechanism Involved in Parkinson’s Disease. Parkinsons Dis. 2018, 2018, 1590465. [Google Scholar] [CrossRef] [PubMed]
  35. Infante, J.; Prieto, C.; Sierra, M.; Sanchez-Juan, P.; Gonzalez-Aramburu, I.; Sanchez-Quintana, C.; Berciano, J.; Combarros, O.; Sainz, J. Comparative blood transcriptome analysis in idiopathic and LRRK2 G2019S-associated Parkinson’s disease. Neurobiol. Aging 2016, 38, 214.e1–214.e5. [Google Scholar] [CrossRef] [PubMed]
  36. Riley, B.E.; Gardai, S.J.; Emig-Agius, D.; Bessarabova, M.; Ivliev, A.E.; Schule, B.; Alexander, J.; Wallace, W.; Halliday, G.M.; Langston, J.W.; et al. Systems-based analyses of brain regions functionally impacted in Parkinson’s disease reveals underlying causal mechanisms. PLoS ONE 2014, 9, e102909. [Google Scholar] [CrossRef] [PubMed]
  37. Durrenberger, P.F.; Grunblatt, E.; Fernando, F.S.; Monoranu, C.M.; Evans, J.; Riederer, P.; Reynolds, R.; Dexter, D.T. Inflammatory Pathways in Parkinson’s Disease; A BNE Microarray Study. Parkinsons Dis. 2012, 2012, 214714. [Google Scholar] [CrossRef]
  38. Zhang, B.; Xia, C.; Lin, Q.; Huang, J. Identification of key pathways and transcription factors related to Parkinson disease in genome wide. Mol. Biol. Rep. 2012, 39, 10881–10887. [Google Scholar] [CrossRef] [PubMed]
  39. Edwards, Y.J.; Beecham, G.W.; Scott, W.K.; Khuri, S.; Bademci, G.; Tekin, D.; Martin, E.R.; Jiang, Z.; Mash, D.C.; Ffrench-Mullen, J.; et al. Identifying consensus disease pathways in Parkinson’s disease using an integrative systems biology approach. PLoS ONE 2011, 6, e16917. [Google Scholar] [CrossRef]
  40. Grunblatt, E.; Mandel, S.; Jacob-Hirsch, J.; Zeligson, S.; Amariglo, N.; Rechavi, G.; Li, J.; Ravid, R.; Roggendorf, W.; Riederer, P.; et al. Gene expression profiling of parkinsonian substantia nigra pars compacta; alterations in ubiquitin-proteasome, heat shock protein, iron and oxidative stress regulated proteins, cell adhesion/cellular matrix and vesicle trafficking genes. J. Neural Transm. 2004, 111, 1543–1573. [Google Scholar] [CrossRef]
  41. Mandel, S.; Grunblatt, E.; Riederer, P.; Amariglio, N.; Jacob-Hirsch, J.; Rechavi, G.; Youdim, M.B. Gene expression profiling of sporadic Parkinson’s disease substantia nigra pars compacta reveals impairment of ubiquitin-proteasome subunits, SKP1A, aldehyde dehydrogenase, and chaperone HSC-70. Ann. N. Y. Acad. Sci. 2005, 1053, 356–375. [Google Scholar] [PubMed]
  42. Bogetofte, H.; Ryan, B.J.; Jensen, P.; Schmidt, S.I.; Vergoossen, D.L.E.; Barnkob, M.B.; Kiani, L.N.; Chughtai, U.; Heon-Roberts, R.; Caiazza, M.C.; et al. Post-translational proteomics platform identifies neurite outgrowth impairments in Parkinson’s disease GBA-N370S dopamine neurons. Cell Rep. 2023, 42, 112180. [Google Scholar] [CrossRef] [PubMed]
  43. Jang, Y.; Pletnikova, O.; Troncoso, J.C.; Pantelyat, A.Y.; Dawson, T.M.; Rosenthal, L.S.; Na, C.H. Mass Spectrometry-Based Proteomics Analysis of Human Substantia Nigra From Parkinson’s Disease Patients Identifies Multiple Pathways Potentially Involved in the Disease. Mol. Cell Proteom. 2023, 22, 100452. [Google Scholar]
  44. Acera, A.; Gomez-Esteban, J.C.; Murueta-Goyena, A.; Galdos, M.; Azkargorta, M.; Elortza, F.; Ruzafa, N.; Ibarrondo, O.; Pereiro, X.; Vecino, E. Potential Tear Biomarkers for the Diagnosis of Parkinson’s Disease-A Pilot Study. Proteomes 2022, 10, 4. [Google Scholar] [CrossRef] [PubMed]
  45. Zafar, S.; Noor, A.; Younas, N.; Shafiq, M.; Schmitz, M.; Wurster, I.; Brockmann, K.; Gasser, T.; Zerr, I. SWATH Mass Spectrometry-Based CSF Proteome Profile of GBA-Linked Parkinson’s Disease Patients. Int. J. Mol. Sci. 2022, 23, 14166. [Google Scholar] [CrossRef] [PubMed]
  46. Zhao, Y.; Zhang, Y.; Zhang, J.; Yang, G. Plasma proteome profiling using tandem mass tag labeling technology reveals potential biomarkers for Parkinson’s disease: A preliminary study. Proteom. Clin. Appl. 2022, 16, e2100010. [Google Scholar] [CrossRef] [PubMed]
  47. Jiang, R.; Rong, C.; Ke, R.; Meng, S.; Yan, X.; Ke, H.; Wu, S. Differential proteomic analysis of serum exosomes reveals alterations in progression of Parkinson disease. Medicine 2019, 98, e17478. [Google Scholar] [CrossRef] [PubMed]
  48. Sandor, C.; Honti, F.; Haerty, W.; Szewczyk-Krolikowski, K.; Tomlinson, P.; Evetts, S.; Millin, S.; Keane, T.; McCarthy, S.A.; Durbin, R.; et al. Whole-exome sequencing of 228 patients with sporadic Parkinson’s disease. Sci. Rep. 2017, 7, 41188. [Google Scholar] [CrossRef]
  49. Hu, Y.; Deng, L.; Zhang, J.; Fang, X.; Mei, P.; Cao, X.; Lin, J.; Wei, Y.; Zhang, X.; Xu, R. A Pooling Genome-Wide Association Study Combining a Pathway Analysis for Typical Sporadic Parkinson’s Disease in the Han Population of Chinese Mainland. Mol. Neurobiol. 2016, 53, 4302–4318. [Google Scholar] [CrossRef]
  50. O’Dushlaine, C.; Kenny, E.; Heron, E.A.; Segurado, R.; Gill, M.; Morris, D.W.; Corvin, A. The SNP ratio test: Pathway analysis of genome-wide association datasets. Bioinformatics 2009, 25, 2762–2763. [Google Scholar] [CrossRef]
  51. Schapira, A.H.V.; Chaudhuri, K.R.; Jenner, P. Non-motor features of Parkinson disease. Nat. Rev. Neurosci. 2017, 18, 435–450. [Google Scholar] [CrossRef]
  52. Jellinger, K.A. Synuclein deposition and non-motor symptoms in Parkinson disease. J. Neurol. Sci. 2011, 310, 107–111. [Google Scholar] [CrossRef] [PubMed]
  53. Bang, D.; Lim, S.; Lee, S.; Kim, S. Biomedical knowledge graph learning for drug repurposing by extending guilt-by-association to multiple layers. Nat. Commun. 2023, 14, 3570. [Google Scholar] [CrossRef]
  54. Nalls, M.A.; Blauwendraat, C.; Vallerga, C.L.; Heilbron, K.; Bandres-Ciga, S.; Chang, D.; Tan, M.; Kia, D.A.; Noyce, A.J.; Xue, A.; et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 2019, 18, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  55. Foo, J.N.; Chew, E.G.Y.; Chung, S.J.; Peng, R.; Blauwendraat, C.; Nalls, M.A.; Mok, K.Y.; Satake, W.; Toda, T.; Chao, Y.; et al. Identification of Risk Loci for Parkinson Disease in Asians and Comparison of Risk Between Asians and Europeans: A Genome-Wide Association Study. JAMA Neurol. 2020, 77, 746–754. [Google Scholar] [CrossRef]
  56. Chang, D.; Nalls, M.A.; Hallgrimsdottir, I.B.; Hunkapiller, J.; van der Brug, M.; Cai, F.; International Parkinson’s Disease Genomics Consortium; 23andMe Research Team; Kerchner, G.A.; Ayalon, G.; et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat. Genet. 2017, 49, 1511–1516. [Google Scholar] [CrossRef] [PubMed]
  57. Tsou, P.S.; Sawalha, A.H. Glycoprotein nonmetastatic melanoma protein B: A key mediator and an emerging therapeutic target in autoimmune diseases. FASEB J. 2020, 34, 8810–8823. [Google Scholar] [CrossRef] [PubMed]
  58. Tripathi, U.; Rosh, I.; Ben Ezer, R.; Nayak, R.; Chowdhary, A.; Djamus, J.; Manole, A.; Haulden, H.; Gage, F.H.; Stern, S. Upregulated extracellular matrix-related genes and impaired synaptic activity in dopaminergic and hippocampal neurons derived from Parkinson’s disease patients with PINK1 and PARK2 mutations. bioRxiv 2022. [Google Scholar] [CrossRef]
  59. Rosh, I.; Tripathi, U.; Hussein, Y.; Rike, W.A.; Manole, A.; Cordeiro, D.; Houlden, H.; Winkler, J.; Gage, F.; Stern, S. Synaptic dysfunction and dysregulation of extracellular matrix-related genes in dopaminergic neurons derived from Parkinson’s disease sporadic patients and with GBA1 mutations. bioRxiv 2023. [Google Scholar] [CrossRef]
  60. Rike, W.A.; Stern, S. Proteins and Transcriptional Dysregulation of the Brain Extracellular Matrix in Parkinson’s Disease: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 7435. [Google Scholar] [CrossRef]
  61. Teves, J.M.Y.; Bhargava, V.; Kirwan, K.R.; Corenblum, M.J.; Justiniano, R.; Wondrak, G.T.; Anandhan, A.; Flores, A.J.; Schipper, D.A.; Khalpey, Z.; et al. Parkinson’s Disease Skin Fibroblasts Display Signature Alterations in Growth, Redox Homeostasis, Mitochondrial Function, and Autophagy. Front. Neurosci. 2017, 11, 737. [Google Scholar] [CrossRef] [PubMed]
  62. Gene Ontology Browser. Extracelullar Matrix Organization. Available online: https://www.informatics.jax.org/vocab/gene_ontology/GO:0030198# (accessed on 15 October 2023).
  63. Liu, C.Z.; Guo, D.S.; Ma, J.J.; Dong, L.R.; Chang, Q.Q.; Yang, H.Q.; Liang, K.K.; Li, X.H.; Yang, D.W.; Fan, Y.Y.; et al. Correlation of matrix metalloproteinase 3 and matrix metalloproteinase 9 levels with non-motor symptoms in patients with Parkinson’s disease. Front. Aging Neurosci. 2022, 14, 889257. [Google Scholar] [CrossRef] [PubMed]
  64. Downs, M.; Sethi, M.K.; Raghunathan, R.; Layne, M.D.; Zaia, J. Matrisome changes in Parkinson’s disease. Anal. Bioanal. Chem. 2022, 414, 3005–3015. [Google Scholar] [CrossRef] [PubMed]
  65. Freitas, A.; Aroso, M.; Rocha, S.; Ferreira, R.; Vitorino, R.; Gomez-Lazaro, M. Bioinformatic analysis of the human brain extracellular matrix proteome in neurodegenerative disorders. Eur. J. Neurosci. 2021, 53, 4016–4033. [Google Scholar] [CrossRef] [PubMed]
  66. Gupta, V.; Singh, M.K.; Garg, R.K.; Pant, K.K.; Khattri, S. Evaluation of peripheral matrix metalloproteinase-1 in Parkinson’s disease: A case-control study. Int. J. Neurosci. 2014, 124, 88–92. [Google Scholar] [CrossRef] [PubMed]
  67. Farkas, E.; De Jong, G.I.; de Vos, R.A.; Jansen Steur, E.N.; Luiten, P.G. Pathological features of cerebral cortical capillaries are doubled in Alzheimer’s disease and Parkinson’s disease. Acta Neuropathol. 2000, 100, 395–402. [Google Scholar] [CrossRef]
  68. Downs, M.; Zaia, J.; Sethi, M.K. Mass spectrometry methods for analysis of extracellular matrix components in neurological diseases. Mass. Spectrom. Rev. 2023, 42, 1848–1875. [Google Scholar] [CrossRef] [PubMed]
  69. Rey, F.; Pandini, C.; Messa, L.; Launi, R.; Barzaghini, B.; Zangaglia, R.; Raimondi, M.T.; Gagliardi, S.; Cereda, C.; Zuccotti, G.V.; et al. alpha-Synuclein antisense transcript SNCA-AS1 regulates synapses- and aging-related genes suggesting its implication in Parkinson’s disease. Aging Cell 2021, 20, e13504. [Google Scholar] [CrossRef]
  70. Huang, Y.; Wen, D.; Yuan, Y.; Chen, W. Gene Set Enrichment Analysis and Genetic Experiment Reveal Changes in Cell Signaling Pathways Induced by alpha-Synuclein Overexpression. Biomedicines 2023, 11, 263. [Google Scholar] [CrossRef]
  71. Mehra, S.; Sahay, S.; Maji, S.K. alpha-Synuclein misfolding and aggregation: Implications in Parkinson’s disease pathogenesis. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 890–908. [Google Scholar] [CrossRef]
  72. Riederer, P.; Berg, D.; Casadei, N.; Cheng, F.; Classen, J.; Dresel, C.; Jost, W.; Krüger, R.; Müller, T.; Reichmann, H.; et al. alpha-Synuclein in Parkinson’s disease: Causal or bystander? J. Neural Transm. 2019, 126, 815–840. [Google Scholar] [CrossRef] [PubMed]
  73. Roy, A.; Chalapathi, A.V.; Balagurunathan, K. Investigating the Roles of Heparan Sulfate Structures in Alpha-Synuclein Aggregation in Cell Culture Models. Methods Mol. Biol. 2022, 2303, 807–820. [Google Scholar] [PubMed]
  74. Mehra, S.; Ghoshn, D.; Kumar, R.; Mondal, M.; Gadhe, L.G.; Das, S.; Anoop, A.; Jha, N.N.; Jacob, R.S.; Chatterjee, D.; et al. Glycosaminoglycans have variable effects on alpha-synuclein aggregation and differentially affect the activities of the resulting amyloid fibrils. J. Biol. Chem. 2018, 293, 12975–12991. [Google Scholar] [CrossRef] [PubMed]
  75. Maiza, A.; Chantepie, S.; Vera, C.; Fifre, A.; Huynh, M.B.; Stettler, O.; Ouidja, M.O.; Papy-Garcia, D. The role of heparan sulfates in protein aggregation and their potential impact on neurodegeneration. FEBS Lett. 2018, 592, 3806–3818. [Google Scholar] [CrossRef] [PubMed]
  76. Ihse, E.; Yamakado, H.; van Wijk, X.M.; Lawrence, R.; Esko, J.D.; Masliah, E. Cellular internalization of alpha-synuclein aggregates by cell surface heparan sulfate depends on aggregate conformation and cell type. Sci. Rep. 2017, 7, 9008. [Google Scholar] [CrossRef] [PubMed]
  77. Adulla, A.; Patel, U.; Ashok, A.; Katiyar, P.; Kaulakis, M.; Kritikos, A.E.; Pillai, S.; Lee, H.; Lindner, E.; Rhee, D.J.; et al. Alpha-Synuclein modulates fibronectin expression in the trabecular meshwork independent of TGFbeta2. Exp. Eye Res. 2023, 226, 109351. [Google Scholar] [CrossRef] [PubMed]
  78. Paiva, I.; Jain, G.; Lazaro, D.F.; Jercic, K.G.; Hentrich, T.; Kerimoglu, C.; Pinho, R.; Szegő, M.; Burkhardt, S.; Capece, V.; et al. Alpha-synuclein deregulates the expression of COL4A2 and impairs ER-Golgi function. Neurobiol. Dis. 2018, 119, 121–135. [Google Scholar] [CrossRef] [PubMed]
  79. Cohlberg, J.A.; Li, J.; Uversky, V.N.; Fink, A.L. Heparin and other glycosaminoglycans stimulate the formation of amyloid fibrils from alpha-synuclein in vitro. Biochemistry 2002, 41, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
  80. Ju, C.; Gao, J.; Hou, L.; Wang, L.; Zhang, F.; Sun, F.; Zhang, T.; Xu, P.; Shi, Z.; Hu, F.; et al. Neuroprotective effect of chondroitin sulfate on SH-SY5Y cells overexpressing wild-type or A53T mutant alpha-synuclein. Mol. Med. Rep. 2017, 16, 8721–8728. [Google Scholar] [CrossRef]
  81. Tsuboi, K.; Grzesiak, J.J.; Bouvet, M.; Hashimoto, M.; Masliah, E.; Shults, C.W. Alpha-synuclein overexpression in oligodendrocytic cells results in impaired adhesion to fibronectin and cell death. Mol. Cell Neurosci. 2005, 29, 259–268. [Google Scholar] [CrossRef]
  82. Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Garbis, S.D.; Ntzouni, M.; Margaritis, L.H.; Stefanis, L.; Vekrellis, K. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851. [Google Scholar] [CrossRef]
  83. Yamada, K.; Iwatsubo, T. Extracellular alpha-synuclein levels are regulated by neuronal activity. Mol. Neurodegener. 2018, 13, 9. [Google Scholar] [CrossRef]
  84. Ferreira, S.A.; Romero-Ramos, M. Microglia Response During Parkinson’s Disease: Alpha-Synuclein Intervention. Front. Cell Neurosci. 2018, 12, 247. [Google Scholar] [CrossRef]
  85. Surguchev, A.A.; Surguchov, A. Integrins-A missing link in synuclein’s pathogenic mechanism. J. Neurosci. Res. 2019, 97, 539–542. [Google Scholar] [CrossRef] [PubMed]
  86. Hou, L.; Bao, X.; Zang, C.; Yang, H.; Sun, F.; Che, Y.; Wu, X.; Li, S.; Zhang, D.; Wang, Q. Integrin CD11b mediates alpha-synuclein-induced activation of NADPH oxidase through a Rho-dependent pathway. Redox Biol. 2018, 14, 600–608. [Google Scholar] [CrossRef] [PubMed]
  87. Li, J.Y.; Englund, E.; Holton, J.L.; Soulet, D.; Hagell, P.; Lees, A.J.; Lashley, T.; Quinn, N.P.; Rehncrona, S.; Björklund, A.; et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat. Med. 2008, 14, 501–503. [Google Scholar] [CrossRef]
  88. DeWitt, D.A.; Richey, P.L.; Praprotnik, D.; Silver, J.; Perry, G. Chondroitin sulfate proteoglycans are a common component of neuronal inclusions and astrocytic reaction in neurodegenerative diseases. Brain Res. 1994, 656, 205–209. [Google Scholar] [CrossRef]
  89. Lehri-Boufala, S.; Ouidja, M.O.; Barbier-Chassefiere, V.; Henault, E.; Raisman-Vozari, R.; Garrigue-Antar, L.; Papy-Garcia, D.; Morin, C. New roles of glycosaminoglycans in alpha-synuclein aggregation in a cellular model of Parkinson disease. PLoS ONE 2015, 10, e0116641. [Google Scholar] [CrossRef] [PubMed]
  90. Tao, Y.; Sun, Y.; Lv, S.; Xia, W.; Zhao, K.; Xu, Q.; Zhao, Q.; He, L.; Le, W.; Wang, Y.; et al. Heparin induces alpha-synuclein to form new fibril polymorphs with attenuated neuropathology. Nat. Commun. 2022, 13, 4226. [Google Scholar] [CrossRef]
  91. Nikolaus, S.; Antke, C.; Muller, H.W. In vivo imaging of synaptic function in the central nervous system: I. Movement disorders and dementia. Behav. Brain Res. 2009, 204, 1–31. [Google Scholar] [CrossRef]
  92. Sousa, V.L.; Bellani, S.; Giannandrea, M.; Yousuf, M.; Valtorta, F.; Meldolesi, J.; Chieregatti, E. {alpha}-synuclein and its A30P mutant affect actin cytoskeletal structure and dynamics. Mol. Biol. Cell 2009, 20, 3725–3739. [Google Scholar] [CrossRef] [PubMed]
  93. Dankovich, T.M.; Rizzoli, S.O. The Synaptic Extracellular Matrix: Long-Lived, Stable, and Still Remarkably Dynamic. Front. Synaptic Neurosci. 2022, 14, 854956. [Google Scholar] [CrossRef] [PubMed]
  94. Ruffini, N.; Klingenberg, S.; Schweiger, S.; Gerber, S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020, 9, 2642. [Google Scholar] [CrossRef] [PubMed]
  95. Li, P.; Nie, Y.; Yu, J. An Effective Method to Identify Shared Pathways and Common Factors among Neurodegenerative Diseases. PLoS ONE 2015, 10, e0143045. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, Y.; Sakaguchi, M.; Sabit, H.; Tamai, S.; Ichinose, T.; Tanaka, S.; Kinoshita, M.; Uchida, Y.; Ohtsuki, S.; Nakada, M. COL1A2 inhibition suppresses glioblastoma cell proliferation and invasion. J. Neurosurg. 2023, 138, 639–648. [Google Scholar] [CrossRef] [PubMed]
  97. Brennan, S.; Keon, M.; Liu, B.; Su, Z.; Saksena, N.K. Panoramic Visualization of Circulating MicroRNAs Across Neurodegenerative Diseases in Humans. Mol. Neurobiol. 2019, 56, 7380–7407. [Google Scholar] [CrossRef] [PubMed]
  98. Hirsch, E.C. Biochemistry of Parkinson’s disease with special reference to the dopaminergic systems. Mol. Neurobiol. 1994, 9, 135–142. [Google Scholar] [CrossRef]
  99. Aguila, J.; Cheng, S.; Kee, N.; Cao, M.; Wang, M.; Deng, Q.; Hedlund, E. Spatial RNA Sequencing Identifies Robust Markers of Vulnerable and Resistant Human Midbrain Dopamine Neurons and Their Expression in Parkinson’s Disease. Front. Mol. Neurosci. 2021, 14, 699562. [Google Scholar] [CrossRef] [PubMed]
  100. Hynes, R.O. The extracellular matrix: Not just pretty fibrils. Science 2009, 326, 1216–1219. [Google Scholar] [CrossRef]
  101. Thalhammer, A.; Cingolani, L.A. Cell adhesion and homeostatic synaptic plasticity. Neuropharmacology 2014, 78, 23–30. [Google Scholar] [CrossRef]
  102. Dityatev, A.; Seidenbecher, C.I.; Schachner, M. Compartmentalization from the outside: The extracellular matrix and functional microdomains in the brain. Trends Neurosci. 2010, 33, 503–512. [Google Scholar] [CrossRef]
  103. Soles, A.; Selimovic, A.; Sbrocco, K.; Ghannoum, F.; Hamel, K.; Moncada, E.L.; Gilliat, S.; Cvetanovic, M. Extracellular Matrix Regulation in Physiology and in Brain Disease. Int. J. Mol. Sci. 2023, 24, 7049. [Google Scholar] [CrossRef]
  104. Schulz-Schaeffer, W.J. The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson’s disease and Parkinson’s disease dementia. Acta Neuropathol. 2010, 120, 131–143. [Google Scholar] [CrossRef] [PubMed]
  105. Plowey, E.D.; Chu, C.T. Synaptic dysfunction in genetic models of Parkinson’s disease: A role for autophagy? Neurobiol. Dis. 2011, 43, 60–67. [Google Scholar] [CrossRef] [PubMed]
  106. Ng, X.Y.; Cao, M. Dysfunction of synaptic endocytic trafficking in Parkinson’s disease. Neural Regen. Res. 2024, 19, 2649–2660. [Google Scholar] [CrossRef] [PubMed]
  107. Dityatev, A.; Schachner, M.; Sonderegger, P. The dual role of the extracellular matrix in synaptic plasticity and homeostasis. Nat. Rev. Neurosci. 2010, 11, 735–746. [Google Scholar] [CrossRef]
  108. Mehdi, S.J.; Rosas-Hernandez, H.; Cuevas, E.; Lantz, S.M.; Barger, S.W.; Sarkar, S.; Paule, M.G.; Ali, S.F.; Imam, S.Z. Protein Kinases and Parkinson’s Disease. Int. J. Mol. Sci. 2016, 17, 1585. [Google Scholar] [CrossRef] [PubMed]
  109. Kingwell, K. LRRK2-targeted Parkinson disease drug advances into phase III. Nat. Rev. Drug Discov. 2023, 22, 3–5. [Google Scholar] [CrossRef]
  110. Chapman, M.A. Interactions between cell adhesion and the synaptic vesicle cycle in Parkinson’s disease. Med. Hypotheses 2014, 83, 203–207. [Google Scholar] [CrossRef]
  111. Bao, Y.; Wang, L.; Liu, H.; Yang, J.; Yu, F.; Cui, C.; Huang, D. A Diagnostic Model for Parkinson’s Disease Based on Anoikis-Related Genes. Mol. Neurobiol. 2024, 61, 3641–3656. [Google Scholar] [CrossRef]
  112. Pantaleo, E.; Monaco, A.; Amoroso, N.; Lombardi, A.; Bellantuono, L.; Urso, D.; Giudice, C.L.; Picardi, E.; Tafuri, B.; Nigro, S.; et al. A Machine Learning Approach to Parkinson’s Disease Blood Transcriptomics. Genes 2022, 13, 727. [Google Scholar] [CrossRef]
  113. Uehara, Y.; Ueno, S.I.; Amano-Takeshige, H.; Suzuki, S.; Imamichi, Y.; Fujimaki, M.; Ota, N.; Murase, T.; Inoue, T.; Saiki, S.; et al. Non-invasive diagnostic tool for Parkinson’s disease by sebum RNA profile with machine learning. Sci. Rep. 2021, 11, 18550. [Google Scholar] [CrossRef]
  114. Odumpatta, R.; Arumugam, M. Integrative Analysis of Gene Expression and Regulatory Network Interaction Data Reveals the Protein Kinase C Family of Serine/Threonine Receptors as a Significant Druggable Target for Parkinson’s Disease. J. Mol. Neurosci. 2021, 71, 466–480. [Google Scholar] [CrossRef]
  115. Hu, L.; Dong, M.X.; Huang, Y.L.; Lu, C.Q.; Qian, Q.; Zhang, C.C.; Xu, X.M.; Liu, Y.; Chen, G.H.; Wei, Y.D. Integrated Metabolomics and Proteomics Analysis Reveals Plasma Lipid Metabolic Disturbance in Patients With Parkinson’s Disease. Front. Mol. Neurosci. 2020, 13, 80. [Google Scholar] [CrossRef]
  116. Yuan, Q.; Zhang, S.; Li, J.; Xiao, J.; Li, X.; Yang, J.; Lu, D.; Wang, Y. Comprehensive analysis of core genes and key pathways in Parkinson’s disease. Am. J. Transl. Res. 2020, 12, 5630–5639. [Google Scholar]
  117. Moni, M.A.; Rana, H.K.; Islam, M.B.; Ahmed, M.B.; Xu, H.; Hasan, M.A.M.; Lei, Y.; Quinn, J.M. A computational approach to identify blood cell-expressed Parkinson’s disease biomarkers that are coordinately expressed in brain tissue. Comput. Biol. Med. 2019, 113, 103385. [Google Scholar] [CrossRef]
  118. Dong, N.; Zhang, X.; Liu, Q. Identification of therapeutic targets for Parkinson’s disease via bioinformatics analysis. Mol. Med. Rep. 2017, 15, 731–735. [Google Scholar] [CrossRef]
  119. Rezaei-Tavirani, M.; Zamanian-Azodi, M.; Rajabi, S.; Masoudi-Nejad, A.; Rostami-Nejad, M.; Rahmatirad, S. Protein Clustering and Interactome Analysis in Parkinson and Alzheimer’s Diseases. Arch. Iran. Med. 2016, 19, 101–109. [Google Scholar]
  120. Dumitriu, A.; Golji, J.; Labadorf, A.T.; Gao, B.; Beach, T.G.; Myers, R.H.; Longo, K.A.; Latourelle, J.C. Integrative analyses of proteomics and RNA transcriptomics implicate mitochondrial processes, protein folding pathways and GWAS loci in Parkinson disease. BMC Med. Genom. 2016, 9, 5. [Google Scholar] [CrossRef] [PubMed]
  121. Calligaris, R.; Banica, M.; Roncaglia, P.; Robotti, E.; Finaurini, S.; Vlachouli, C.; Antonutti, L.; Iorio, F.; Carissimo, A.; Cattaruzza, T.; et al. Blood transcriptomics of drug-naive sporadic Parkinson’s disease patients. BMC Genom. 2015, 16, 876. [Google Scholar] [CrossRef] [PubMed]
  122. Hao, B.; Chen, X.; Dai, D.; Zou, C.; Wu, X.; Chen, J. Bioinformatic analysis of microRNA expression in Parkinson’s disease. Mol. Med. Rep. 2015, 11, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  123. Licker, V.; Cote, M.; Lobrinus, J.A.; Rodrigo, N.; Kovari, E.; Hochstrasser, D.F.; Turck, N.; Sanchez, J.C.; Burkhard, P.R. Proteomic profiling of the substantia nigra demonstrates CNDP2 overexpression in Parkinson’s disease. J. Proteom. 2012, 75, 4656–4667. [Google Scholar] [CrossRef] [PubMed]
  124. Botta-Orfila, T.; Sanchez-Pla, A.; Fernandez, M.; Carmona, F.; Ezquerra, M.; Tolosa, E. Brain transcriptomic profiling in idiopathic and LRRK2-associated Parkinson’s disease. Brain Res. 2012, 1466, 152–157. [Google Scholar] [CrossRef] [PubMed]
  125. Shi, M.; Jin, J.; Wang, Y.; Beyer, R.P.; Kitsou, E.; Albin, R.L.; Gearing, M.; Pan, C.; Zhang, J. Mortalin: A protein associated with progression of Parkinson disease? J. Neuropathol. Exp. Neurol. 2008, 67, 117–124. [Google Scholar] [CrossRef]
  126. Ferraro, F.; Fevga, C.; Bonifati, V.; Mandemakers, W.; Mahfouz, A.; Reinders, M. Correcting Differential Gene Expression Analysis for Cyto-Architectural Alterations in Substantia Nigra of Parkinson’s Disease Patients Reveals Known and Potential Novel Disease-Associated Genes and Pathways. Cells 2022, 11, 198. [Google Scholar] [CrossRef] [PubMed]
  127. Kurvits, L.; Lattekivi, F.; Reimann, E.; Kadastik-Eerme, L.; Kasterpalu, K.M.; Koks, S.; Taba, P.; Planken, A. Transcriptomic profiles in Parkinson’s disease. Exp. Biol. Med. 2021, 246, 584–595. [Google Scholar] [CrossRef]
  128. Licker, V.; Turck, N.; Kovari, E.; Burkhardt, K.; Cote, M.; Surini-Demiri, M.; Lobrinus, J.A.; Sanchez, J.C.; Burkhard, P.R. Proteomic analysis of human substantia nigra identifies novel candidates involved in Parkinson’s disease pathogenesis. Proteomics 2014, 14, 784–794. [Google Scholar] [CrossRef] [PubMed]
  129. Lopez-Cerdan, A.; Andreu, Z.; Hidalgo, M.R.; Grillo-Risco, R.; Catala-Senent, J.F.; Soler-Saez, I.; Neva-Alejo, A.; Gordillo, F.; de la Iglesia-Vayá, M.; García-García, F. Unveiling sex-based differences in Parkinson’s disease: A comprehensive meta-analysis of transcriptomic studies. Biol. Sex. Differ. 2022, 13, 68. [Google Scholar] [CrossRef]
  130. Riboldi, G.M.; Vialle, R.A.; Navarro, E.; Udine, E.; de Paiva Lopes, K.; Humphrey, J.; Allan, A.; Parks, M.; Henderson, B.; Astudillo, K.; et al. Transcriptome deregulation of peripheral monocytes and whole blood in GBA-related Parkinson’s disease. Mol. Neurodegener. 2022, 17, 52. [Google Scholar] [CrossRef]
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta Analyses (PRISMA) flow diagram outlining the search strategy.
Figure 1. Preferred Reporting Items for Systematic Reviews and Meta Analyses (PRISMA) flow diagram outlining the search strategy.
Brainsci 14 00522 g001
Figure 2. Transcriptomic studies: Top deregulated ECM-related gene sets/pathways. Columns are grouped by adhesion (gray), ECM or collagen (light green), GAG related (light yellow), and MMP related (light blue). Large circles indicate that the gene set or pathway was among the top 5 and smaller circles indicate that the gene set or pathway was among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways, green circles denote GO gene sets, and yellow circles denote Reactome or other pathways [17,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. Abbreviations: ECM = extracellular matrix, GAG = glycosaminoglycan, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes, MMP = matrix metalloprotease, org = organization, proc = process.
Figure 2. Transcriptomic studies: Top deregulated ECM-related gene sets/pathways. Columns are grouped by adhesion (gray), ECM or collagen (light green), GAG related (light yellow), and MMP related (light blue). Large circles indicate that the gene set or pathway was among the top 5 and smaller circles indicate that the gene set or pathway was among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways, green circles denote GO gene sets, and yellow circles denote Reactome or other pathways [17,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. Abbreviations: ECM = extracellular matrix, GAG = glycosaminoglycan, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes, MMP = matrix metalloprotease, org = organization, proc = process.
Brainsci 14 00522 g002
Figure 3. Proteomics studies: Top deregulated ECM-related gene sets/pathways. Columns are grouped by adhesion (gray), ECM or collagen (light green), GAG-proteoglycan related (light yellow), and integrin signaling (light blue). Large circles indicate gene sets or pathways among the top 5, and small circles among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways, green circles denote GO gene sets, and yellow circles denote Reactome or other pathways [18,36,42,43,44,45,46,47]. Abbreviations: ECM = extracellular matrix, GAG = glycosaminoglycan, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes, metab = metabolism, proc = process.
Figure 3. Proteomics studies: Top deregulated ECM-related gene sets/pathways. Columns are grouped by adhesion (gray), ECM or collagen (light green), GAG-proteoglycan related (light yellow), and integrin signaling (light blue). Large circles indicate gene sets or pathways among the top 5, and small circles among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways, green circles denote GO gene sets, and yellow circles denote Reactome or other pathways [18,36,42,43,44,45,46,47]. Abbreviations: ECM = extracellular matrix, GAG = glycosaminoglycan, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes, metab = metabolism, proc = process.
Brainsci 14 00522 g003
Figure 4. Genomics studies: Top enriched ECM-related gene sets/pathways. Columns are grouped by adhesion (gray) and ECM (light green). Large circles indicate gene sets or pathways among the top 5, and small circles among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways and green circles denote GO gene sets [39,48,49,50]. Abbreviations: ECM = extracellular matrix, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes.
Figure 4. Genomics studies: Top enriched ECM-related gene sets/pathways. Columns are grouped by adhesion (gray) and ECM (light green). Large circles indicate gene sets or pathways among the top 5, and small circles among the top 10, as defined by individual study authors. Dark-blue circles denote KEGG pathways and green circles denote GO gene sets [39,48,49,50]. Abbreviations: ECM = extracellular matrix, GO = gene ontology, KEGG = Kyoto Encyclopedia of Genes and Genomes.
Brainsci 14 00522 g004
Table 1. Summary of included studies.
Table 1. Summary of included studies.
Authors, YearTissueMethod
Transcriptomics Studies
Rosh et al., 2024 [26]iPSC-derived DA neuronsRNA sequencing
Akrioti et al., 2022 [27]iPSC-derived neural progenitor cellsRNA sequencing
Stern et al., 2022 [28]iPSC-derived DA neuronsRNA sequencing
Hemmings et al., 2022 [29]BloodRNA sequencing
Cho et al., 2021 [30]MSCs from adipose tissueRNA sequencing
Fernandez-Santiago et al., 2021 [17]Dermal fibroblastsRNA sequencing
Yang et al., 2021 [31]BloodRNA sequencing
Booth et al., 2019 [32]iPSC-derived, midbrain-patterned astrocytes generated from skinRNA sequencing
Gonzalez-Cascuberta et al., 2018 [33]Dermal fibroblastsRNA sequencing
Tan et al., 2018 [34]BloodMicroarray
Infante et al., 2016 [35]Whole bloodRNA sequencing
Riley et al., 2014 [36]Substantia nigra, striatum, cortexMicroarray
Durrenberger et al., 2012 [37]Substantia nigraMicroarray
Zhang et al., 2012 [38]Substantia nigraMicroarray
Edwards et al., 2011 [39]Dorsal motor nucleus of vagus, locus coeruleus, substantia nigra, putamen, insulaMicroarray
Grunblatt et al., 2004 [40];
Mandel et al., 2005 [41]
Substantia nigra, pars compactaMicroarray
Proteomics studies
Bogetofte et al., 2023 [42]iPSC-derived DA neuronsLC/MS
Jang et al., 2023 [43]Substantia nigraLC/MS
Acera et al., 2022 [44]TearsLC/MS
Zafar et al., 2022 [45]Cerebrospinal fluidLC/MS
Zhao et al., 2022 [46]PlasmaLC/MS
Raghunathan et al., 2020 [18]Prefrontal cortexLC/MS
Jiang et al., 2019 [47]Serum exosomesLC/MS
Riley et al., 2014 [36]Striatum, cortexLC/MS
Genomics studies
Sandor et al., 2017 [48]Not reportedWhole exome sequencing
Hu et al., 2016 [49]White blood cellsGWAS
Edwards et al., 2011 [39]Not reportedGWAS
O’Dushlaine et al., 2009 [50]Not reportedGWAS/SNP ratio test
Abbreviations: DA = dopamine, GWAS = genome wide association study, iPSC = induced pluripotential stem cells, LC/MS = liquid chromatography/mass spectrometry, MSC = mesenchymal stem cells, SNP = single nucleotide polymorphism.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chapman, M.A.; Sorg, B.A. A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson’s Disease. Brain Sci. 2024, 14, 522. https://doi.org/10.3390/brainsci14060522

AMA Style

Chapman MA, Sorg BA. A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson’s Disease. Brain Sciences. 2024; 14(6):522. https://doi.org/10.3390/brainsci14060522

Chicago/Turabian Style

Chapman, Mary Ann, and Barbara A. Sorg. 2024. "A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson’s Disease" Brain Sciences 14, no. 6: 522. https://doi.org/10.3390/brainsci14060522

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop