Next Article in Journal
The Impact of Hydroxychloroquine on Primary Feto-Placental Endothelial Cells from Healthy and Early-Onset Preeclamptic Placentas
Next Article in Special Issue
Mitochondrial Dynamics in Neurodegenerative Diseases: Unraveling the Role of Fusion and Fission Processes
Previous Article in Journal
Efficacy of Green Synthesized Nanoparticles in Photodynamic Therapy: A Therapeutic Approach
Previous Article in Special Issue
GATA3 as a Blood-Based RNA Biomarker for Idiopathic Parkinson’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Brain-Biomarker Changes in Body Fluids of Patients with Parkinson’s Disease

Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10932; https://doi.org/10.3390/ijms241310932
Submission received: 22 May 2023 / Revised: 20 June 2023 / Accepted: 27 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue Peripheral Biomarkers in Neurodegenerative Diseases 3.0)

Abstract

:
Parkinson’s disease (PD) is an incurable neurodegenerative disease that is rarely diagnosed at an early stage. Although the understanding of PD-related mechanisms has greatly improved over the last decade, the diagnosis of PD is still based on neurological examination through the identification of motor symptoms, including bradykinesia, rigidity, postural instability, and resting tremor. The early phase of PD is characterized by subtle symptoms with a misdiagnosis rate of approximately 16–20%. The difficulty in recognizing early PD has implications for the potential use of novel therapeutic approaches. For this reason, it is important to discover PD brain biomarkers that can indicate early dopaminergic dysfunction through their changes in body fluids, such as saliva, urine, blood, or cerebrospinal fluid (CSF). For the CFS-based test, the invasiveness of sampling is a major limitation, whereas the other body fluids are easier to obtain and could also allow population screening. Following the identification of the crucial role of alpha-synuclein (α-syn) in the pathology of PD, a very large number of studies have summarized its changes in body fluids. However, methodological problems have led to the poor diagnostic/prognostic value of this protein and alternative biomarkers are currently being investigated. The aim of this paper is therefore to summarize studies on protein biomarkers that are alternatives to α-syn, particularly those that change in nigrostriatal areas and in biofluids, with a focus on blood, and, eventually, saliva and urine.

1. Introduction

1.1. PD

PD is a brain disorder with an irreversible and continuous process of neurodegeneration until death. There is no cure for this disease, but treatments are available, of which levodopa (L-dopa) is the gold standard. The disease is diagnosed by neurologists by observing motor symptoms such as bradykinesia, rigidity, postural instability, and resting tremor [1,2]. However, all these symptoms occur years (possibly decades) after the onset of the neurodegenerative process, leading to difficulties in recognizing early PD, which is characterized by hyposmia, depression, constipation, sleep disturbances (REM-sleep-behavior disorder, excessive daytime sleepiness) [3,4]. As a result, misdiagnosis rates are in the range of 10–30% [5,6,7], which reduces the therapeutic benefit and compromises the possibility of using novel therapeutic approaches. For this reason, significant effort is underway in the search for candidate biomarkers that can track disease progression and/or differentiate PD from the other neurodegenerative diseases, as well as being able to detect early changes at the level of the basal ganglia. The basal ganglia are a group of subcortical nuclei including the striatum (STR), which is subdivided into dorsal (caudate and putamen) and ventral (nucleus accumbens), the globus pallidus (external and internal parts), the subthalamic nucleus, and the substantia nigra (SN) [8]. The SN is divided into two areas: the pars compacta (SNpc), which consists mainly of dopaminergic perikarya, and the pars reticulata (SNpr), which consists mainly of gamma-aminobutyric acid (GABA)-inhibitory neurons. The SNpc has dopaminergic projections to the striatum, putamen, and caudate nuclei. Within the striatum, these dopaminergic projections synapse on the D1- and D2-family receptor neurons, which are involved in the “direct and indirect pathways” of the basal ganglia [9]. Furthermore, PD is not only characterized by alterations in the dopaminergic system, but the map of altered substances includes several neuroproteins, neurotransmitters, and neuropeptides that are expressed or not expressed within the nigro-striatal circuits [10,11,12].

1.2. PD Biomarkers: General Information

Because PD is a multifactorial disease, several types of biomarkers are under study, including clinical, brain-based (such as neuroimaging), genetic, and biochemical. Clinical biomarkers, such as bradykinesia, resting tremor, and muscle rigidity are used by clinicians to identify PD and monitor both response to medical treatment and disease progression. However, other clinical features, such as hyposmia, rapid eye movement (REM) sleep-behavior disorder (RBD), and constipation are under consideration as biomarkers of prodromal PD [3,4]. Neuroimaging techniques, such as transcranial sonography, magnetic resonance imaging (MRI), positron-emission tomography (PET) or single-photon-emission computed tomography (SPECT) are considered biomarkers of nigrostriatal neurodegeneration and may also detect the early phase of PD and follow its progression [13]. Genetic biomarkers are considered important tools in the diagnosis of PD, and a family history is reported in approximately 10–20% of PD patients [14]. Of the various genes identified as biomarkers, some are associated with autosomal recessive inheritance, particularly Parkin and PINK1, which are associated with typical early-onset PD [15]; others are related to atypical juvenile PD, such as ATP13A2, DNAJC6, FBOX7, and SYNJ1 [16], whereas SNCA, LRRK2, and VPS35 are associated with typical autosomal dominant PD [17]. In general, studies of genetic risk factors for PD have been limited to single-nucleotide variants, which account for a small fraction of the genetic variation in the human genome. Recent results from researchers involved in the PD Biomarker Programme (PDBP: https://pdbp.ninds.nih.gov/news, accessed on 6 June 2023) identified a structural-genotype variant associated with PD risk in 2585 patients compared with 2779 controls [18]. In terms of biochemical biomarkers, there are a large number of studies focusing on different biomolecules present in the brain and/or body fluids, including metabolites as glutathione, pyruvate, and glycine derivatives [19], plasma lipids such as N-acylphosphatidyl ethanolamines [20], and a large number of proteins, including α-syn. There are several methods used to evaluate biochemical biomarkers. Quantitative reverse-transcription PCR (RT-qPCR) or in situ hybridization can be used to identify the mRNA, while electronic, fluorescence and confocal microscopy may be useful to investigate where a molecule is expressed and/or in which cell types. However, the most useful techniques for detecting biomarkers in the blood include multicolor flow cytometry, the radioimmunoassay (RIA), the enzyme-linked immunosorbent assay (ELISA), the single-molecule array (Simoa), and chemiluminescence. In the last decade, high-performance liquid chromatography–mass spectrometry (HPLC-MS) has become an essential part of biomarker-discovery research through targeted, semi-targeted or non-targeted measurement analysis, allowing large amounts of data to be collected with high accuracy from a variety of samples, including bodily fluids. Interestingly, through a spectrometry-based method, the analysis of SN from PD patients and controls in a PDBP study allowed the identification of several mechanisms leading to dopaminergic cell death and α-syn deposition, including those related to ribosome activity and GABAergic synapse [21]. Following the identification of the crucial role of brain α-syn in the pathology of PD, a very large number of studies summarized the changes to this protein in body fluids, particularly in the CSF. However, methodological problems have led to the poor diagnostic/prognostic value of this protein, as shown in several studies [22,23,24], including one carried out in a large Parkinson’s Progression Markers Initiative (PPMI) cohort of patients [25], and alternative biomarkers are currently under investigation. The aim of this paper is, therefore, to focus on brain-protein biomarkers, alternatives to α-syn and, in particular, those that may indicate early dopaminergic dysfunction through their changes in bodily fluids. With regard to CSF, despite its close association with the brain, the invasiveness of sampling is a limitation, whereas the other bodily fluids are easier to obtain and could also allow population screening, so the present review focuses on blood and, eventually, saliva and urine. To the author’s knowledge, reviews on brain-protein biomarkers that change in the biofluids are rare. These candidate biomarkers are reviewed for their potential to aid in early diagnosis, prognosis, or monitoring of specific clinical features.

2. Brain-Biomarker Changes in Body Fluids of PD Patients

2.1. Neurofilament Light Protein

Neurofilaments (NFs) are members of a family of intermediate filament proteins characterized by an ‘intermediate’ diameter (10 nm), which is larger than that of actin filaments (6 nm) and smaller than that of myosin filaments (15 nm) [26], and include NF-L, -M, and -H, corresponding to light, medium, and heavy, respectively [27]. These NF proteins occupy the axonal cytoplasm, support neurons, and interact with several other proteins that regulate synaptogenesis and neurotransmission [28,29]. The -L, -M, and -H NFs were found by immunohistochemistry (IHC) to be abnormally aggregated within Lewy bodies in the post-mortem SN of a PD patient (whose disease stage was not reported) [30]. Blood NF-L levels measured by ELISA were used to discriminate PD from atypical parkinsonism [31] with the same diagnostic accuracy as CSF NF-L [32]. In addition, plasma NF-L was correlated with disease severity and the progression of both motor and cognitive functions using an electrochemiluminescence immunoassay [33], while when the serum NF-L was measured by Simoa together with other fluid biomarkers, it helped to discriminate PD patients from controls [34]. In a complete longitudinal PPMI cohort study, a large dataset of NF-L measurements by Simoa or ELISA was obtained from: (i) 176 CSF samples, including newly diagnosed and drug-naïve PD patients and controls, (ii) 1190 sera from patients with PD (including newly diagnosed and drug-naïve patients) (iii) other neurodegenerative diseases, (iv) subjects with prodromal conditions and mutation carriers as well as sera form healthy controls, [35]. The results showed that the serum and CSF NF-L levels were significantly correlated and were highest in other neurodegenerative diseases, but higher in the PD patients than in the controls. Furthermore, the NF-L levels increased over time and with age (by 3.35% per year of age, and women had a median serum level 6.79%, which was higher than that of the men), were correlated with PD severity and, most importantly, were highest in early PD, but also relatively high in the prodromal groups, including patients affected by RBD disorders, suggesting a prognostic role.
Similar results were obtained in another large study [36], in which PD patients were selected on the basis of diagnosis within 2 years, not receiving treatment at baseline, Hoehn and Yahr (H-Y) stage 1 or 2, 123I-isoflurane DaT imaging, and, to avoid misdiagnosis, a longitudinal review of their diagnosis. The study included CSF samples from 207 PD patients and 102 controls, while the sera were collected from 361 PD patients and 176 controls, plus 291 individuals who provided both serum and CSF samples. Using Simoa, the baseline CSF and serum NF-L levels were both higher in the PD patients than in the controls, but the CSF NF-L levels were significantly elevated in the males compared to the females, which is not consistent with the study cited above. In addition, the same study showed in a cross-sectional and prospective follow-up of the PD patients that baseline CSF and serum NF-L levels could predict motor decline and tremor (but not rigidity). Interestingly serum NF-L levels measured by Simoa may also be useful to differentiate PD from essential tremor (ET) [37]. Indeed, when NF-L levels were measured by electrochemiluminescence immunoassay in sera from patients with PD (>2 years of diagnosis; n = 146), ET (n = 82) and 60 age-matched healthy controls, significantly higher levels were found in PD than in ET and healthy controls. In the same study, serum NF-L was found to be elevated in patients with advanced H-Y stage and dementia and was an independent contributor to motor symptoms and cognitive severity. In 289 patients with advanced PD, NF-L measured by Simoa was associated with motor function, cognitive decline, and subclinical cardiac damage [38]. A study focusing on the comparability of PPMI and Asian cohorts highlighted the importance of adjusting several variables, including demographics, age, and sex, to best interpret serum NF-L levels for research and clinical practice [39]. However, despite all these important findings, the diagnostic accuracy of serum NF-L is low [35] and changes are not specific for PD [40,41,42,43]. Therefore, rather than being a diagnostic tool for PD, NF-L may be a useful marker for certain specific aspects of PD, such as dementia, cognitive decline, or cardiac damage. It may also be a good biomarker for distinguishing PD from ET.

2.2. Substance P

Substance P (Sub-P) is an 11-amino-acid peptide that belongs to the tachykinin family and is derived from a pre-protachykinin polyprotein precursor of the TAC1 gene [44,45]. Sub-P acts through various mechanisms, including autocrine, paracrine, and endocrine, to modulate pain perception by altering cellular signaling pathways [46,47]. Sub-P mRNA is found in the GABAergic neuron terminals of the SN [48], where the protein acts to sensitize postsynaptic dopaminergic neurons to glutamate [49]. In humans, the presence of Sub-P within α-syn aggregates was demonstrated by IHC in the perikarya of the colon [50] and olfactory bulb [51]. Sub-P changes in SN and/or STR of PD patients have been extensively studied in both animal models and patients. Sub-P levels detected by IHC and/or RIA were decreased in the SN and STR of both unilateral and bilateral rats injected with 6-hydroxydopamine (6-OHDA) [52,53,54], whereas the opposite results were obtained in rats treated with intraperitoneal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [55,56]. Consistent with animal models, when human post-mortem brains were measured using various techniques, including RIA, reduced levels of Sub-P were found in the SN [57,58,59], globus pallidus [59], putamen [58,60], and caudate [61] of PD patients compared with control brain samples. In patients with early (n = 15) and advanced (n = 15) PD, sputum Sub-P levels were assessed by ELISA and were significantly decreased in the advanced-stage patients compared to the controls and early-stage patients [62]. In another study [63] including 22 PD patients (with a score of 2 on the H-Y scale and treated with L-dopa) and 12 controls, serum Sub-P levels were measured by competitive ELISA. The Sub-P levels were higher than in the controls and increased proportionally with the disease severity (motor impairment). The limited number of available studies on Sub-P in the bodily fluids of PD patients and the small number of subjects involved make it difficult to identify Sub-P as a reliable blood (or saliva) biomarker. However, the presence of Sub-P within α-syn aggregates needs to be investigated further, especially for its possible involvement in early PD.

2.3. S100 Calcium-Binding Protein A10

The S100s are a family of low-molecular-weight proteins with two calcium-binding sites that have a helix–loop–helix conformation. Approximately 21 different proteins have been recognized and are encoded by the S100A1, S100A2, and S100A3 genes (among others) genes [64]. The S100 calcium-binding protein A10 (namely S100A10 or P11), encoded by the S100A10 gene, modulates neuronal function [65]. The expression of P11 was investigated in post-mortem brains (both the SN and the putamen from fresh snap-frozen samples) of PD patients (3 females and 2 males) and controls (4 females and 1 male). The P11 levels, which were submitted to a Western blot (WB) analysis, decreased in the dopaminergic SN cells, along with the mRNA detected by laser-capture microdissection coupled to quantitative real-time PCR [66]. In the same study, P11 was also investigated by multicolor flow cytometry in leukocytes from a total of 42 PD patients with or without depression (at different disease stages, including de novo PD patients and those with H-Y scores of) and 15 controls. The P11 levels in the CD8+ cells were found to be higher in the depressed and non-depressed PD patients compared to the controls (with a sensitivity and specificity of 93%), but increased exclusively in the CD14+CD16− of the depressed PD patients, corresponding to the same modulation reported in depressed patients without PD [67]. Furthermore, the P11 levels in the monocytes, cytotoxic T cells, and NK cells were all correlated with PD severity. Although the pathological role of P11 in PD remains unclear, as with its role in normal SN, P11 can be considered as a PD biomarker that is useful in identifying patients at risk of developing depression [68].

2.4. Neurotensin

Neurotensin (NT) is a 13-amino-acid peptide [69] that is widely distributed in neurons, in which, in dense nuclear vesicles, it acts as a neuromodulator of several neurotransmitters, such as acetylcholine, as well as serotonin, GABA, and dopamine [70].The IHC of normal rat brains showed the expression of NT in ventral tegmental-area fibers and SN (both compacta and reticulata) [71], while combined HPLC/RIA showed an increased (twofold) expression in post-mortem SN samples from 6 advanced-PD patients (compared to 5 controls) [72]. In another study, using specific RIA on postmortem brain samples from 25 human patients with advanced PD compared to controls, the NT levels were significantly decreased in the hippocampus but not altered in the SN [73]. Using RIA on blood samples from 36 subjects (16 controls,16 patients with advanced disease, and 4 untreated patients), the NT concentrations were higher in the PD patients than in the controls and higher in the untreated patients than in the patients treated with L-dopa [74]. However, the number of studies using blood from early PD patients is excessively small, so further studies are needed to clarify the role of NT as a blood biomarker in PD.

2.5. Chromogranins

Chromogranin proteins [75] are the major components of secretory granules in neuroendocrine cells, where they play a role in granulogenesis and in the sorting and processing of secretory-protein cargo [76]. Chromogranin proteins are considered biomarkers of neuroendocrine neoplasia [77]. Many proteins belong to the chromogranin family, including chromogranins A (CgA) and B (CgB) and secretogranin II (SgII, or chromogranin C) [78]. Several studies have reported that chromogranin-family proteins are found in the SN and are involved in PD changes. One study reported that CgA, detected by IHC with an antibody against the WE14 epitope, was expressed in non-dopaminergic neurons of the SNpc [79]. Processed and unprocessed forms of CgA were expressed in anatomically defined GABAergic, glutamatergic, cholinergic, and catecholaminergic neurons in the rodent central nervous system [79]. In human studies, the midbrain, cerebellum, and cerebrum of 7 PD patients and 2 PD-associated Alzheimer’s disease (AD) patients were examined by light and electron microscopy using antibodies against synaptophysin and ChA, and compared with 10 controls. Lewy bodies were observed in the hematoxylin- and eosin-stained sections of all the PD and PD/AD cases. Within the Lewy bodies, the antibodies mainly stained the peripheral zones, and at the ultrastructural level, labeling was found in the vesicular, filamentous, and granular structures. However, the antibodies did not show any differences between the PD and normal controls on immunoblot analysis [80]. In another study, post-mortem brains from 4 cases of AD, 2 cases of Pick’s disease, 4 cases of PD and various other diseases, and brains from 6 cases without neurological disorders were examined for CgA expression by IHC. Lewy bodies with vesicles in the periphery of the central nucleus were immunoreactive for CgA in the SN of the PD brain samples but also in those with other diseases [81]. Another study [82] showed that serum levels of chromogranins and secretogranins were correlated with the progression and severity of the PD. The subjects included in the study were patients with early (n = 14), intermediate (n = 18), or late (n = 4) stages of PD, according to their H-Y scores, as well as well-defined PD (n = 36) and healthy controls (n = 52). The serum concentrations of CgA, CgB, and SgII were measured by ELISA. Compared with the controls, the serum CgA levels were significantly increased and the serum SgII levels were significantly decreased in the overall population of PD patients, whereas the serum CgB levels did not differ between the two groups. In the early-stage group, the CgA and SgII levels were lower and higher, respectively, than in the other groups. Both the CgA and the SgII serum levels changed progressively over time and were correlated with both the H-Y and the UPDRS scores. The results for the chromogranins were promising, but the number of early-stage PD patients studied was small, and it remains to be investigated whether the blood changes are characteristic of PD or of neurodegenerative diseases in general, as has been observed using CSF [83].

2.6. VGF

The VGF gene (not abbreviated) is regulated by the nerve-growth factor (NGF) in PC12 cells and cultured cortical neurons [84]. The VGF precursor (proVGF), consisting of 617/615 aa in rats/mice and humans, respectively, with >85% identity (minor sequence differences between rat and human) [85], is expressed in rat brains [86], but also in human plasma [87,88]. It is induced by growth factors [89] and can give rise to a variety of truncated peptides, including the so-called TLQP [90,91,92], NERP1, and NERP2 [93]. Peptide sequences of VGF have mainly been identified in CSF, where specific peptide alterations have been associated with certain diseases. For example, VGF peptides cleaved at the N-terminus of proVGF were found to be altered in CSF of psychiatric disorders [94,95,96,97], while others in the middle of proVGF may be useful CSF biomarkers for dementia [98,99,100]. The VGF immunostaining (using an antibody against the last nine amino acids at the C-terminus) was decreased in GABAergic (containing Sub-P) fibers of the SN in rats treated with 6-OHDA, but it was restored by L-dopa treatment [101]. Unfortunately, VGF expression in human SN remains to be investigated, but VGF alterations of certain peptides (TPGH and NERP-1) have been found in the post-mortem parietal cortexes of PD patients [102]. In addition, the next-generation RNA sequencing of cingulate cortex samples (from 23 PD patients, compared with 11 controls) found the VGF gene to be implicated in the molecular etiology of PD and PD-related dementia [103]. The levels of VGF C-terminal (C-t) peptides were analyzed by a home-made competitive ELISA using an antibody directed against the last nine amino acids at the C-terminus of human proVGF in PD patients at the time of diagnosis (drug-free, n = 23) or after dopamine replacement (n = 40) compared to age-matched controls (n = 21) [101]. In the drug-free patients, a strong decrease (>50%) was observed at the time of diagnosis, whereas long-term L-dopa treatment caused an increase in VGF. The levels of VGF C-t peptides were also correlated with disease duration, LED, and the severity of olfactory dysfunction, but not with the H-Y score. The VGF changes observed in the blood of PD patients were verified in other bodily fluids, such as the CSF and urine. When CSF samples were analyzed from two independent cohorts of subjects (n = 196) and a longitudinal cohort (n = 105), including all the controls and treated PD patients (with disease duration of no more than 6 years), the C-terminal region of the VGF protein was found to be decreased by liquid chromatography–tandem mass spectrometry in the data-independent acquisition mode [104]. Using the same technique, more than 200 urine samples from different groups of subjects were analyzed, including (i) healthy controls, (ii) non-manifest carriers of the LRRK2 G2019S mutation, (iii) idiopathic PD patients, and (iv) manifest PD patients with LRRK2 G2019S. The results showed that changes (decrease) were found in the peptides covering most of the VGF sequence [105]. Interestingly, CSF and urine data were found to be correlated [106]. Decreased levels of VGF C-t peptides have also been studied in plasma samples from amyotrophic lateral sclerosis patients, but a small reduction was found only in the late stage of the disease [87] while no changes in plasma-VGF C-t peptides were observed in a rat model of schizophrenia [107]. In conclusion, VGF C-t peptides are potential blood biomarkers for PD, as they appear to be selectively altered in PD. However, both the use of a more specific sandwich ELISA and confirmation by HPLC-MS are required to accurately identify individual VGF sequences that are altered in PD blood.

2.7. Glial Fibrillary Acidic Protein

Glial fibrillary acidic protein (GFAP) is an intermediate filament protein found primarily in mature astrocytes. This protein has a much broader function than the provision of mechanical support to cells, as it is involved in the production and regulation of the blood–brain barrier, increases synaptic plasticity, and coordinates neuronal activity [108]. This protein is upregulated in brain damage or neuronal degeneration; therefore, it is considered a marker of astrocyte activation (or astrogliosis) [109]. Increased numbers of GFAP-immunoreactive astrocytes were observed in the SN of mice after only one day of MPTP administration [110], as well as after 6-OHDA treatment [111]. The role of astrogliosis in the PD brain is not fully elucidated, although most of the relevant studies observed slight or no increases in the expression of both GFAP-immunoreactive cells (determined by IHC-stereological quantification and WB analysis) in postmortem samples from SN collected from patients with PD [112,113,114]. Only Damier et al. observed the opposite results in their study, i.e., the increased expression (according to their IHC analysis) of GFAP in the SN of PD patients correlated with the percentage of lost dopaminergic neurons [115]. Changes in GFAP levels were also studied in the plasma of PD patients. Tang et al. observed an increase in GFAP levels (using Simoa) in PD patients (n = 60) compared to healthy controls (n = 15), and the highest increase was in PD patients with dementia (n = 24) [116]. Similar results were obtained with serum samples in which an increase in GFAP levels (detected by sandwich ELISA) was observed in PD patients (n = 23) compared with healthy controls (n = 29) [117] and, again, this increase was more pronounced in PD patients with mild cognitive impairment or dementia than in PD patients with normal cognition [118]. Another cross-sectional study examined PD patients characterized by RBD, an important risk factor for cognitive impairment in PD patients [119]. These patients (n = 39) had higher serum GFAP levels compared to PD patients without RBD (n = 70) and healthy controls (n = 37, measured with Simoa) [120], suggesting the role of GFAP as a predictive PD biomarker. Elevated serum levels of this protein are also observed in patients with other neurodegenerative diseases, such as AD and frontotemporal lobar degeneration [121,122].

3. Conclusions

PD is one of the most significant neurodegenerative diseases, and because most of the cardinal symptoms occur late in its course, a large number of dopaminergic neurons are already damaged when it is diagnosed. Therefore, there is a great need for biomarkers that can be used to ensure early diagnoses, as well as to differentiate PD from other diseases with high sensitivity and specificity, and to track disease progression. When biomarkers are altered in both the brain and body fluids it is more likely that changes in the periphery are linked to the changes occurring in the central nervous system. In addition, improvements in the identification of the neuronal proteins that change in PD could lead to a better understanding of the PD pathological mechanisms, which in turn could lead to new approaches to diagnosis and treatment. Although the candidate biomarkers studied here are promising (Table 1 and Figure 1), they have limitations related to (i) the small number of blood samples, especially in the early PD, (ii) their low specificity in PD, (iii) the lack of studies based on orthogonal methods, which are often required to provide the independent confirmation of the results. At this point, it is important to note that a single biomarker is unlikely to achieve the higher standard of sensitivity and specificity required for an accurate diagnosis of PD, as the mechanisms involved in PD are very complex and sometimes similar to those in other neurodegenerative diseases. Therefore, another good strategy may be to validate a large panel of PD fluid biomarkers, including those non-specific that may increase in value when combined.

Funding

This research received no external funding.

Institutional Review Board Statement

This study did not require ethical approval because it reports others studies.

Informed Consent Statement

Informed consent was obtained from all subjects involved in each study reported in this review.

Data Availability Statement

This is a review, the authors reported other published studies, hence they have not the permission to share the results.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Przedborski, S. The Two-Century Journey of Parkinson Disease Research. Nat. Rev. Neurosci. 2017, 18, 251–259. [Google Scholar] [CrossRef] [PubMed]
  2. Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s Disease. J. Neural. Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef] [PubMed]
  3. Goldman, J.G.; Postuma, R. Premotor and Nonmotor Features of Parkinson’s Disease. Curr. Opin. Neurol. 2014, 27, 434–441. [Google Scholar] [CrossRef] [Green Version]
  4. Siderowf, A.; Lang, A.E. Premotor Parkinson’s Disease: Concepts and Definitions. Mov. Disord. 2012, 27, 608–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hess, C.W.; Okun, M.S. Diagnosing Parkinson Disease. Contin. Lifelong Learn. Neurol. 2016, 22, 1047–1063. [Google Scholar] [CrossRef]
  6. Joutsa, J.; Gardberg, M.; Röyttä, M.; Kaasinen, V. Diagnostic Accuracy of Parkinsonism Syndromes by General Neurologists. Park. Relat. Disord. 2014, 20, 840–844. [Google Scholar] [CrossRef]
  7. Poewe, W.; Wenning, G. The Differential Diagnosis of Parkinson’s Disease. Eur. J. Neurol. 2002, 9, 23–30. [Google Scholar] [CrossRef]
  8. del Rey, N.L.-G.; García-Cabezas, M.Á. Cytology, Architecture, Development, and Connections of the Primate Striatum: Hints for Human Pathology. Neurobiol. Dis. 2023, 176, 105945. [Google Scholar] [CrossRef]
  9. Afifi, A.K. Topical Review: Basal Ganglia: Functional Anatomy and Physiology. Part 2. J. Child. Neurol. 1994, 9, 352–361. [Google Scholar] [CrossRef]
  10. Emamzadeh, F.N.; Surguchov, A. Parkinson’s Disease: Biomarkers, Treatment, and Risk Factors. Front. Neurosci. 2018, 12, 612. [Google Scholar] [CrossRef] [Green Version]
  11. Lotankar, S.; Prabhavalkar, K.S.; Bhatt, L.K. Biomarkers for Parkinson’s Disease: Recent Advancement. Neurosci. Bull. 2017, 33, 585–597. [Google Scholar] [CrossRef] [PubMed]
  12. Zheng, Y.; Zhang, L.; Xie, J.; Shi, L. The Emerging Role of Neuropeptides in Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 646726. [Google Scholar] [CrossRef] [PubMed]
  13. Mitchell, T.; Lehéricy, S.; Chiu, S.Y.; Strafella, A.P.; Stoessl, A.J.; Vaillancourt, D.E. Emerging Neuroimaging Biomarkers Across Disease Stage in Parkinson Disease. JAMA Neurol. 2021, 78, 1262. [Google Scholar] [CrossRef] [PubMed]
  14. Farrer, M.J. Genetics of Parkinson Disease: Paradigm Shifts and Future Prospects. Nat. Rev. Genet. 2006, 7, 306–318. [Google Scholar] [CrossRef]
  15. Brooks, J.; Ding, J.; Simon-Sanchez, J.; Paisan-Ruiz, C.; Singleton, A.B.; Scholz, S.W. Parkin and PINK1 Mutations in Early-Onset Parkinson’s Disease: Comprehensive Screening in Publicly Available Cases and Control. J. Med. Genet. 2009, 46, 375–381. [Google Scholar] [CrossRef]
  16. Wittke, C.; Petkovic, S.; Dobricic, V.; Schaake, S.; Arzberger, T.; Compta, Y.; Englund, E.; Ferguson, L.W.; Gelpi, E.; Roeber, S.; et al. Genotype–Phenotype Relations for the Atypical Parkinsonism Genes: MDSGene Systematic Review. Mov. Disord. 2021, 36, 1499–1510. [Google Scholar] [CrossRef]
  17. Lesage, S.; Houot, M.; Mangone, G.; Tesson, C.; Bertrand, H.; Forlani, S.; Anheim, M.; Brefel-Courbon, C.; Broussolle, E.; Thobois, S.; et al. Genetic and Phenotypic Basis of Autosomal Dominant Parkinson’s Disease in a Large Multi-Center Cohort. Front. Neurol. 2020, 11, 682. [Google Scholar] [CrossRef]
  18. Billingsley, K.J.; Ding, J.; Jerez, P.A.; Illarionova, A.; Levine, K.; Grenn, F.P.; Makarious, M.B.; Moore, A.; Vitale, D.; Reed, X.; et al. Genome-Wide Analysis of Structural Variants in Parkinson Disease. Ann. Neurol. 2023, 93, 1012–1022. [Google Scholar] [CrossRef]
  19. Havelund, J.; Heegaard, N.; Færgeman, N.; Gramsbergen, J. Biomarker Research in Parkinson’s Disease Using Metabolite Profiling. Metabolites 2017, 7, 42. [Google Scholar] [CrossRef] [Green Version]
  20. Hamid, Z.; Basit, A.; Pontis, S.; Piras, F.; Assogna, F.; Bossù, P.; Pontieri, F.E.; Stefani, A.; Spalletta, G.; Franceschi, P.; et al. Gender Specific Decrease of a Set of Circulating N-Acylphosphatidyl Ethanolamines (NAPEs) in the Plasma of Parkinson’s Disease Patients. Metabolomics 2019, 15, 74. [Google Scholar] [CrossRef] [Green Version]
  21. 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] [CrossRef]
  22. Ganguly, U.; Singh, S.; Pal, S.; Prasad, S.; Agrawal, B.K.; Saini, R.V.; Chakrabarti, S. Alpha-Synuclein as a Biomarker of Parkinson’s Disease: Good, but Not Good Enough. Front. Aging Neurosci. 2021, 13, 702639. [Google Scholar] [CrossRef]
  23. Du, T.; Wang, L.; Liu, W.; Zhu, G.; Chen, Y.; Zhang, J. Biomarkers and the Role of α-Synuclein in Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 645996. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, J.; Idowu, A.; Rosenthal, L.; Mao, X. Parkinson’s Disease Fluid Biomarkers for Differential Diagnosis of Atypical Parkinsonian Syndromes. Clin. Transl. Discov. 2023, 3, e150. [Google Scholar] [CrossRef]
  25. Kang, J.-H.; Mollenhauer, B.; Coffey, C.S.; Toledo, J.B.; Weintraub, D.; Galasko, D.R.; Irwin, D.J.; Van Deerlin, V.; Chen-Plotkin, A.S.; Caspell-Garcia, C.; et al. CSF Biomarkers Associated with Disease Heterogeneity in Early Parkinson’s Disease: The Parkinson’s Progression Markers Initiative Study. Acta Neuropathol. 2016, 131, 935–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Schmitt, F.O.; Geren, B.B. The fibrous structure of the nerve axon in relation to the localization of “neurotubules”. J. Exp. Med. 1950, 91, 499–504. [Google Scholar] [CrossRef] [Green Version]
  27. Lee, M.K.; Cleveland, D.W. Neuronal Intermediate Filaments. Annu. Rev. Neurosci. 1996, 19, 187–217. [Google Scholar] [CrossRef]
  28. Yuan, A.; Rao, M.V.; Veeranna; Nixon, R.A. Neurofilaments at a Glance. J. Cell Sci. 2012, 125, 3257–3263. [Google Scholar] [CrossRef] [Green Version]
  29. Sainio, M.T.; Rasila, T.; Molchanova, S.M.; Järvilehto, J.; Torregrosa-Muñumer, R.; Harjuhaahto, S.; Pennonen, J.; Huber, N.; Herukka, S.-K.; Haapasalo, A.; et al. Neurofilament Light Regulates Axon Caliber, Synaptic Activity, and Organelle Trafficking in Cultured Human Motor Neurons. Front. Cell Dev. Biol. 2022, 9, 820105. [Google Scholar] [CrossRef]
  30. Goldman, J.E.; Yen, S.-H.; Chiu, F.-C.; Peress, N.S. Lewy Bodies of Parkinson’s Disease Contain Neurofilament Antigens. Science 1983, 221, 1082–1084. [Google Scholar] [CrossRef]
  31. Marques, T.M.; van Rumund, A.; Oeckl, P.; Kuiperij, H.B.; Esselink, R.A.J.; Bloem, B.R.; Otto, M.; Verbeek, M.M. Serum NFL Discriminates Parkinson Disease from Atypical Parkinsonisms. Neurology 2019, 92, e1479–e1486. [Google Scholar] [CrossRef] [PubMed]
  32. Hansson, O.; Janelidze, S.; Hall, S.; Magdalinou, N.; Lees, A.J.; Andreasson, U.; Norgren, N.; Linder, J.; Forsgren, L.; Constantinescu, R.; et al. Blood-Based NfL: A Biomarker for Differential Diagnosis of Parkinsonian Disorder. Neurology 2017, 88, 930–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lin, C.-H.; Li, C.-H.; Yang, K.-C.; Lin, F.-J.; Wu, C.-C.; Chieh, J.-J.; Chiu, M.-J. Blood NfL: A Biomarker for Disease Severity and Progression in Parkinson Disease. Neurology 2019, 93, e1104–e1111. [Google Scholar] [CrossRef] [PubMed]
  34. Oosterveld, L.P.; Verberk, I.M.W.; Majbour, N.K.; El-Agnaf, O.M.; Weinstein, H.C.; Berendse, H.W.; Teunissen, C.E.; Berg, W.D.J. CSF or Serum Neurofilament Light Added to α-Synuclein Panel Discriminates Parkinson’s from Controls. Mov. Disord. 2020, 35, 288–295. [Google Scholar] [CrossRef] [Green Version]
  35. Mollenhauer, B.; Dakna, M.; Kruse, N.; Galasko, D.; Foroud, T.; Zetterberg, H.; Schade, S.; Gera, R.G.; Wang, W.; Gao, F.; et al. Validation of Serum Neurofilament Light Chain as a Biomarker of Parkinson’s Disease Progression. Mov. Disord. 2020, 35, 1999–2008. [Google Scholar] [CrossRef]
  36. Liu, Y.; Dou, K.; Xue, L.; Li, X.; Xie, A. Neurofilament Light as a Biomarker for Motor Decline in Parkinson’s Disease. Front. Neurosci. 2022, 16, 959261. [Google Scholar] [CrossRef]
  37. Huang, Y.; Huang, C.; Zhang, Q.; Shen, T.; Sun, J. Serum NFL Discriminates Parkinson Disease from Essential Tremor and Reflect Motor and Cognition Severity. BMC Neurol. 2022, 22, 39. [Google Scholar] [CrossRef]
  38. Niemann, L.; Lezius, S.; Maceski, A.; Leppert, D.; Englisch, C.; Schwedhelm, E.; Zeller, T.; Gerloff, C.; Kuhle, J.; Choe, C. Serum Neurofilament Is Associated with Motor Function, Cognitive Decline and Subclinical Cardiac Damage in Advanced Parkinson’s Disease (MARK-PD). Park. Relat. Disord. 2021, 90, 44–48. [Google Scholar] [CrossRef]
  39. Chen, J.-H.; Chan, L.; Chung, C.-C.; Bamodu, O.A.; Hong, C.-T. Blood Neurofilament Light Chain in Parkinson’s Disease: Comparability between Parkinson’s Progression Markers Initiative (PPMI) and Asian Cohorts. J. Clin. Med. 2021, 10, 5085. [Google Scholar] [CrossRef]
  40. Shi, J.; Qin, X.; Chang, X.; Wang, H.; Guo, J.; Zhang, W. Neurofilament Markers in Serum and Cerebrospinal Fluid of Patients with Amyotrophic Lateral Sclerosis. J. Cell Mol. Med. 2022, 26, 583–587. [Google Scholar] [CrossRef]
  41. Zhu, N.; Santos-Santos, M.; Illán-Gala, I.; Montal, V.; Estellés, T.; Barroeta, I.; Altuna, M.; Arranz, J.; Muñoz, L.; Belbin, O.; et al. Plasma Glial Fibrillary Acidic Protein and Neurofilament Light Chain for the Diagnostic and Prognostic Evaluation of Frontotemporal Dementia. Transl. Neurodegener. 2021, 10, 50. [Google Scholar] [CrossRef] [PubMed]
  42. Leuzy, A.; Mattsson-Carlgren, N.; Palmqvist, S.; Janelidze, S.; Dage, J.L.; Hansson, O. Blood-based Biomarkers for Alzheimer’s Disease. EMBO Mol. Med. 2022, 14, e14408. [Google Scholar] [CrossRef] [PubMed]
  43. Disanto, G.; Barro, C.; Benkert, P.; Naegelin, Y.; Schädelin, S.; Giardiello, A.; Zecca, C.; Blennow, K.; Zetterberg, H.; Leppert, D.; et al. Serum Neurofilament Light: A Biomarker of Neuronal Damage in Multiple Sclerosis. Ann. Neurol. 2017, 81, 857–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Steinhoff, M.S.; von Mentzer, B.; Geppetti, P.; Pothoulakis, C.; Bunnett, N.W. Tachykinins and Their Receptors: Contributions to Physiological Control and the Mechanisms of Disease. Physiol. Rev. 2014, 94, 265–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Chang, M.M.; Leeman, S.E.; Niall, H.D. Amino-Acid Sequence of Substance P. Nat. New Biol. 1971, 232, 86–87. [Google Scholar] [CrossRef]
  46. Graefe, S.B.; Mohiuddin, S.S. Biochemistry, Substance P. Available online: https://www.ncbi.nlm.nih.gov/books/NBK554583/?report=printable (accessed on 1 June 2023).
  47. Chang, C.-T.; Jiang, B.-Y.; Chen, C.-C. Ion Channels Involved in Substance P-Mediated Nociception and Antinociception. Int. J. Mol. Sci. 2019, 20, 1596. [Google Scholar] [CrossRef] [Green Version]
  48. Nisbet, A.P.; Foster, O.J.F.; Kingsbury, A.; Eve, D.J.; Daniel, S.E.; Marsden, C.D.; Lees, A.J. Preproenkephalin and Preprotachykinin Messenger RNA Expression in Normal Human Basal Ganglia and in Parkinson’s Disease. Neuroscience 1995, 66, 361–376. [Google Scholar] [CrossRef]
  49. Bolam, J.P.; Smith, Y. The GABA and Substance P Input to Dopaminergic Neurones in the Substantia Nigra of the Rat. Brain Res. 1990, 529, 57–78. [Google Scholar] [CrossRef]
  50. Shannon, K.M.; Keshavarzian, A.; Dodiya, H.B.; Jakate, S.; Kordower, J.H. Is Alpha-Synuclein in the Colon a Biomarker for Premotor Parkinson’s Disease? Evidence from 3 Cases. Mov. Disord. 2012, 27, 716–719. [Google Scholar] [CrossRef]
  51. Smith, R.L.; Baker, H.; Greer, C.A. Immunohistochemical Analyses of the Human Olfactory Bulb. J. Comp. Neurol. 1993, 333, 519–530. [Google Scholar] [CrossRef]
  52. Lindefors, N.; Brodin, E.; Tossman, U.; Segovia, J.; Ungerstedt, U. Tissue Levels and in Vivo Release of Tachykinins and GABA in Striatum and Substantia Nigra of Rat Brain after Unilateral Striatal Dopamine Denervation. Exp. Brain Res. 1989, 74, 527–534. [Google Scholar] [CrossRef]
  53. Salin, P.; Hajji, M.D.; Goff, L. Bilateral 6-Hydroxydopamine-Induced Lesion of the Nigrostriatal Dopamine Pathway Reproduces the Effects of Unilateral Lesion on Substance P but Not on Enkephalin Expression in Rat Basal Ganglia. Eur. Neurosci. Assoc. 1996, 8, 17461–17757. [Google Scholar] [CrossRef]
  54. Taylor, M.D.; De Ceballos, M.L.; Rose, S.; Jenner, P.; Marsden, C.D. Effects of a Unilateral 6-Hydroxydopamine Lesion and Prolonged L-3,4-Dihydroxyphenylalanine Treatment on Peptidergic Systems in Rat Basal Ganglia. Eur. J. Pharmacol. 1992, 219, 183. [Google Scholar] [CrossRef] [PubMed]
  55. Radke, J.M.; Cumming, P.; Vincent, S.R. Effects of MPTP Poisoning on Central Somatostatin and Substance P Levels in the Mouse. Eur. J. Pharmacol. 1987, 134, 105–108. [Google Scholar] [CrossRef] [PubMed]
  56. Meredith, G.E.; Rademacher, D.J. MPTP Mouse Models of Parkinson’s Disease: An Update. J. Park. Dis. 2011, 1, 19–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Clevens, R.A.; Beal, M.F. Substance P-like Immunoreactivity in Brains with Pathological Features of Parkinson’s and Alzheimer’s Diseases. Brain Res. 1989, 486, 387–390. [Google Scholar] [CrossRef]
  58. Tenovuo, O.; Rinne, U.K.; Viljanen, M.K. Substance P Immunoreactivity in the Post-Mortem Parkinsonian Brain. Brain Res. 1984, 303, 113–116. [Google Scholar] [CrossRef]
  59. Mauborgne, A.; Javoy-Agid, F.; Legrand, J.C.; Agid, Y.; Cesselin, F. Decrease of Substance P-like Immunoreactivity in the Substantia Nigra and Pallidum of Parkinsonian Brains. Brain Res. 1983, 268, 167–170. [Google Scholar] [CrossRef]
  60. Sivam, S.P. Dopamine Dependent Decrease in Enkephalin and Substance P Levels in Basal Ganglia Regions of Postmortem Parkinsonian Brains. Neuropeptides 1991, 18, 201–207. [Google Scholar] [CrossRef]
  61. Fernandez, A.; De Ceballos, M.L.; Jenner, P.; Marsden, C.D. Striatal Neuropeptide Levels in Parkinson’s Disease Patients. Neurosci. Lett. 1992, 145, 171–174. [Google Scholar] [CrossRef]
  62. Ebihara, S.; Saito, H.; Kanda, A.; Nakajoh, M.; Takahashi, H.; Arai, H.; Sasaki, H. Impaired Efficacy of Cough in Patients with Parkinson Disease. Chest 2003, 124, 1009–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Schirinzi, T.; Maftei, D.; Ralli, M.; Greco, A.; Mercuri, N.B.; Lattanzi, R.; Severini, C. Serum Substance P Is Increased in Parkinson’s Disease and Correlates with Motor Impairment. Mov. Disord. 2022, 37, 228–230. [Google Scholar] [CrossRef] [PubMed]
  64. Marenholz, I.; Heizmann, C.W.; Fritz, G. S100 Proteins in Mouse and Man: From Evolution to Function and Pathology (Including an Update of the Nomenclature). Biochem. Biophys. Res. Commun. 2004, 322, 1111–1122. [Google Scholar] [CrossRef]
  65. Svenningsson, P.; Greengard, P. P11 (S100A10)—An Inducible Adaptor Protein That Modulates Neuronal Functions. Curr. Opin. Pharmacol. 2007, 7, 27–32. [Google Scholar] [CrossRef] [PubMed]
  66. Green, H.; Zhang, X.; Tiklova, K.; Volakakis, N.; Brodin, L.; Berg, L.; Greengard, P.; Perlmann, T.; Svenningsson, P. Alterations of P11 in Brain Tissue and Peripheral Blood Leukocytes in Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2017, 114, 2735–2740. [Google Scholar] [CrossRef] [Green Version]
  67. Gałecka, M.; Bliźniewska-Kowalska, K.; Gałecki, P.; Szemraj, J.; Orzechowska, A. Expression of P11 in Patients with Depression. J. Clin. Med. 2022, 11, 5743. [Google Scholar] [CrossRef]
  68. Aarsland, D.; Påhlhagen, S.; Ballard, C.G.; Ehrt, U.; Svenningsson, P. Depression in Parkinson Disease—Epidemiology, Mechanisms and Management. Nat. Rev. Neurol. 2012, 8, 35–47. [Google Scholar] [CrossRef]
  69. Carraway, R.; Leeman, S.E. The Isolation of a New Hypotensive Peptide, Neurotensin, from Bovine Hypothalami. J. Biol. Chem. 1973, 248, 6854–6861. [Google Scholar] [CrossRef]
  70. St-Gelais, F.; Jomphe, C.; Trudeau, L.-E. The Role of Neurotensin in Central Nervous System Pathophysiology: What Is the Evidence? J. Psychiatry Neurosci. 2006, 31, 229–245. [Google Scholar]
  71. Vankova, M.; Arluison, M.; Leviel, V.; Tramu, G. Afferent Connections of the Rat Substantia Nigra Pars Lateralis with Special Reference to Peptide-Containing Neurons of the Amygdalo-Nigral Pathway. J. Chem. Neuroanat. 1992, 5, 39–50. [Google Scholar] [CrossRef]
  72. Fernandez, A.; de Ceballos, M.L.; Rose, S.; Jenner, P.; Marsden, C.D. Alterations in Peptide Levels in Parkinson’s Disease and Incidental Lewy Body Disease. Brain 1996, 119, 823–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Bissette, G.; Nemeroff, C.B.; Decker, M.W.; Kizer, J.S.; Agid, Y.; Javoy-Agid, F. Alterations in Regional Brain Concentrations of Neurotensin and Bombesin in Parkinson’s Disease. Ann. Neurol. 1985, 17, 324–328. [Google Scholar] [CrossRef] [PubMed]
  74. Schimpff, R.-M. Increased Plasma Neurotensin Concentrations in Patients with Parkinson’s Disease. J. Neurol. Neurosurg. Psychiatry 2001, 70, 784–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Eiden, L.E.; Huttner, W.B.; Mallet, J.; O’Connor, D.T.; Winkler, H.; Zanini, A. A Nomenclature Proposal for the Chromogranin/Secretogranin Proteins. Neuroscience 1987, 21, 1019–1021. [Google Scholar] [CrossRef]
  76. Estevez-Herrera, J.; Pardo, M.R.; Dominguez, N.; Pereda, D.; Machado, J.D.; Borges, R. The Role of Chromogranins in the Secretory Pathway. Biomol. Concepts 2013, 4, 605–609. [Google Scholar] [CrossRef]
  77. Feldman, S.A.; Eiden, L.E. The Chromogranins: Their Roles in Secretion from Neuroendocrine Cells and as Markers for Neuroendocrine Neoplasia. Endocr. Pathol. 2003, 14, 3–24. [Google Scholar] [CrossRef]
  78. Huttner, W.B.; Gerdes, H.H.; Rosa, P. The Granin-(Chromogranin/Secretogranin) Family. Trends Biochem. Sci. 1991, 16, 27–30. [Google Scholar] [CrossRef]
  79. Schafer, M.K.-H.; Mahata, S.K.; Stroth, N.; Eiden, L.E.; Weihe, E. Cellular Distribution of Chromogranin A in Excitatory, Inhibitory, Aminergic and Peptidergic Neurons of the Rodent Central Nervous System. Regul. Pept. 2010, 165, 36–44. [Google Scholar] [CrossRef] [Green Version]
  80. Nishimura, M.; Tomimoto, H.; Suenaga, T.; Nakamura, S.; Namba, Y.; Ikeda, K.; Akiguchi, I.; Kimura, J. Synaptophysin and Chromogranin A Immunoreactivities of Lewy Bodies in Parkinson’s Disease Brains. Brain Res. 1994, 634, 339–344. [Google Scholar] [CrossRef]
  81. Yasuhara, O.; Kawamata, T.; Aimi, Y.; McGeer, E.G.; McGeer, P.L. Expression of Chromogranin A in Lesions in the Central Nervous System from Patients with Neurological Diseases. Neurosci. Lett. 1994, 170, 13–16. [Google Scholar] [CrossRef]
  82. Xu, D.; Wei, L.; Li, H.; Zhang, W. Serum Levels of Chromogranins and Secretogranins Correlate with the Progress and Severity of Parkinson’s Disease. Kaohsiung J. Med. Sci. 2019, 35, 146–150. [Google Scholar] [CrossRef] [PubMed]
  83. Eder, U.; Leitner, B.; Kirchmair, R.; Pohl, P.; Jobst, K.A.; Smith, A.D.; Mélly, J.; Benzer, A.; Riederer, P.; Reichmann, H.; et al. Levels and Proteolytic Processing of Chromogranin A and B and Secretogranin II in Cerebrospinal Fluid in Neurological Diseases. J. Neural. Transm. 1998, 105, 39–52. [Google Scholar] [CrossRef] [PubMed]
  84. Levi, A.; Eldridge, J.D.; Paterson, B.M. Molecular Cloning of a Gene Sequence Regulated by Nerve Growth Factor. Science 1985, 229, 393–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Canu, N.; Possenti, R.; Ricco, A.S.; Rocchi, M.; Levi, A. Cloning, Structural Organization Analysis, and Chromosomal Assignment of the Human Gene for the Neurosecretory Protein VGF. Genomics 1997, 45, 443–446. [Google Scholar] [CrossRef] [PubMed]
  86. Trani, E.; Giorgi, A.; Canu, N.; Amadoro, G.; Rinaldi, A.M.; Halban, P.A.; Ferri, G.L.; Possenti, R.; Schininà, M.E.; Levi, A. Isolation and Characterization of VGF Peptides in Rat Brain. Role of PC1/3 and PC2 in the Maturation of VGF Precursor. J. Neurochem. 2002, 81, 565–574. [Google Scholar] [CrossRef] [Green Version]
  87. Brancia, C.; Noli, B.; Boido, M.; Boi, A.; Puddu, R.; Borghero, G.; Marrosu, F.; Bongioanni, P.; Orrù, S.; Manconi, B.; et al. VGF Protein and Its C-Terminal Derived Peptides in Amyotrophic Lateral Sclerosis: Human and Animal Model Studies. PLoS ONE 2016, 11, e0164689. [Google Scholar] [CrossRef]
  88. D’Amato, F.; Noli, B.; Angioni, L.; Cossu, E.; Incani, M.; Messana, I.; Manconi, B.; Solinas, P.; Isola, R.; Mariotti, S.; et al. VGF Peptide Profiles in Type 2 Diabetic Patients’ Plasma and in Obese Mice. PLoS ONE 2015, 10, e0142333. [Google Scholar] [CrossRef] [Green Version]
  89. Lewis, J.E.; Brameld, J.M.; Jethwa, P.H. Neuroendocrine Role for VGF. Front. Endocrinol. 2015, 6, 3. [Google Scholar] [CrossRef] [Green Version]
  90. Brancia, C.; Noli, B.; Boido, M.; Pilleri, R.; Boi, A.; Puddu, R.; Marrosu, F.; Vercelli, A.; Bongioanni, P.; Ferri, G.-L.; et al. TLQP Peptides in Amyotrophic Lateral Sclerosis: Possible Blood Biomarkers with a Neuroprotective Role. Neuroscience 2018, 380, 152–163. [Google Scholar] [CrossRef]
  91. Corda, G.; Noli, B.; Manconi, B.; Brancia, C.; Pellegrini, M.; Naro, F.; Olianas, A.; Ferri, G.-L.; Cocco, C. TLQP-21 Changes in Response to a Glucose Load. Tissue Cell 2021, 68, 101471. [Google Scholar] [CrossRef]
  92. Noli, B.; Brancia, C.; Corda, G.; Ferri, G.-L.; Cocco, C. Dynamic of TLQP-Peptides upon Fasting. Tissue Cell 2020, 65, 101368. [Google Scholar] [CrossRef]
  93. D’Amato, F.; Cocco, C.; Noli, B.; Cabras, T.; Messana, I.; Ferri, G.-L. VGF Peptides upon Osmotic Stimuli: Changes in Neuroendocrine Regulatory Peptides 1 and 2 in the Hypothalamic–Pituitary-Axis and Plasma. J. Chem. Neuroanat. 2012, 44, 57–65. [Google Scholar] [CrossRef] [PubMed]
  94. Huang, J.T.-J.; Leweke, F.M.; Oxley, D.; Wang, L.; Harris, N.; Koethe, D.; Gerth, C.W.; Nolden, B.M.; Gross, S.; Schreiber, D.; et al. Disease Biomarkers in Cerebrospinal Fluid of Patients with First-Onset Psychosis. PLoS Med. 2006, 3, e428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Huang, J.T.-J.; Leweke, F.M.; Tsang, T.M.; Koethe, D.; Kranaster, L.; Gerth, C.W.; Gross, S.; Schreiber, D.; Ruhrmann, S.; Schultze-Lutter, F.; et al. CSF Metabolic and Proteomic Profiles in Patients Prodromal for Psychosis. PLoS ONE 2007, 2, e756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jiang, H.; Chen, S.; Lu, N.; Yue, Y.; Yin, Y.; Zhang, Y.; Jiang, W.; Liang, J.; Yuan, Y. Reduced Serum VGF Levels Were Reversed by Antidepressant Treatment in Depressed Patients. World J. Biol. Psychiatry 2017, 18, 586–591. [Google Scholar] [CrossRef]
  97. Li, X.; Ge, H.; Zhou, D.; Wu, X.; Qi, G.; Chen, Z.; Yu, C.; Zhang, Y.; Yu, H.; Wang, C. Reduced Serum VGF Levels Are Linked with Suicide Risk in Chinese Han Patients with Major Depressive Disorder. BMC Psychiatry 2020, 20, 225. [Google Scholar] [CrossRef]
  98. van Steenoven, I.; Noli, B.; Cocco, C.; Ferri, G.L.; Oeckl, P.; Otto, M.; Koel-Simmelink, M.J.A.; Bridel, C.; van der Flier, W.M.; Lemstra, A.W.; et al. VGF Peptides in Cerebrospinal Fluid of Patients with Dementia with Lewy Bodies. Int. J. Mol. Sci. 2019, 20, 4674. [Google Scholar] [CrossRef] [Green Version]
  99. van Steenoven, I.; Koel-Simmelink, M.J.A.; Vergouw, L.J.M.; Tijms, B.M.; Piersma, S.R.; Pham, T.V.; Bridel, C.; Ferri, G.-L.; Cocco, C.; Noli, B.; et al. Identification of Novel Cerebrospinal Fluid Biomarker Candidates for Dementia with Lewy Bodies: A Proteomic Approach. Mol. Neurodegener. 2020, 15, 36. [Google Scholar] [CrossRef]
  100. Boiten, W.; van Steenoven, I.; Xiao, M.-F.; Worley, P.; Noli, B.; Cocco, C.; Ferri, G.-L.; Lemstra, A.; Teunissen, C. Pathologically Decreased CSF Levels of Synaptic Marker NPTX2 in DLB Are Correlated with Levels of Alpha-Synuclein and VGF. Cells 2020, 10, 38. [Google Scholar] [CrossRef]
  101. Cocco, C.; Corda, G.; Lisci, C.; Noli, B.; Carta, M.; Brancia, C.; Manca, E.; Masala, C.; Marrosu, F.; Solla, P.; et al. VGF Peptides as Novel Biomarkers in Parkinson’s Disease. Cell Tissue Res. 2020, 379, 93–107. [Google Scholar] [CrossRef]
  102. Cocco, C.; D’Amato, F.; Noli, B.; Ledda, A.; Brancia, C.; Bongioanni, P.; Ferri, G.L. Distribution of VGF Peptides in the Human Cortex and Their Selective Changes in Parkinson’s and Alzheimer’s Diseases. J. Anat. 2010, 217, 683–693. [Google Scholar] [CrossRef]
  103. Henderson-Smith, A.; Corneveaux, J.J.; De Both, M.; Cuyugan, L.; Liang, W.S.; Huentelman, M.; Adler, C.; Driver-Dunckley, E.; Beach, T.G.; Dunckley, T.L. Next-Generation Profiling to Identify the Molecular Etiology of Parkinson Dementia. Neurol. Genet. 2016, 2, e75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Rotunno, M.S.; Lane, M.; Zhang, W.; Wolf, P.; Oliva, P.; Viel, C.; Wills, A.-M.; Alcalay, R.N.; Scherzer, C.R.; Shihabuddin, L.S.; et al. Cerebrospinal Fluid Proteomics Implicates the Granin Family in Parkinson’s Disease. Sci. Rep. 2020, 10, 2479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Virreira Winter, S.; Karayel, O.; Strauss, M.T.; Padmanabhan, S.; Surface, M.; Merchant, K.; Alcalay, R.N.; Mann, M. Urinary Proteome Profiling for Stratifying Patients with Familial Parkinson’s Disease. EMBO Mol. Med. 2021, 13, e13257. [Google Scholar] [CrossRef] [PubMed]
  106. Karayel, O.; Virreira Winter, S.; Padmanabhan, S.; Kuras, Y.I.; Vu, D.T.; Tuncali, I.; Merchant, K.; Wills, A.M.; Scherzer, C.R.; Mann, M. Proteome Profiling of Cerebrospinal Fluid Reveals Biomarker Candidates for Parkinson’s Disease. Cell Rep. Med. 2022, 3, 100661. [Google Scholar] [CrossRef]
  107. Noli, B.; Sanna, F.; Brancia, C.; D’Amato, F.; Manconi, B.; Vincenzoni, F.; Messana, I.; Melis, M.R.; Argiolas, A.; Ferri, G.-L.; et al. Profiles of VGF Peptides in the Rat Brain and Their Modulations after Phencyclidine Treatment. Front. Cell Neurosci. 2017, 11, 158. [Google Scholar] [CrossRef] [Green Version]
  108. Brenner, M. Role of GFAP in CNS Injuries. Neurosci. Lett. 2014, 565, 7–13. [Google Scholar] [CrossRef] [Green Version]
  109. Middeldorp, J.; Hol, E.M. GFAP in Health and Disease. Prog. Neurobiol. 2011, 93, 421–443. [Google Scholar] [CrossRef]
  110. Muramatsu, Y.; Kurosaki, R.; Watanabe, H.; Michimata, M.; Matsubara, M.; Imai, Y.; Araki, T. Cerebral Alterations in a MPTP-Mouse Model of Parkinson’s Disease—An Immunocytochemical Study. J. Neural. Transm. 2003, 110, 1129–1144. [Google Scholar] [CrossRef]
  111. Gomide, V.C.; Silveira, G.A.; Chadi, G. Transient and Widespread Astroglial Activation in the Brain after a Striatal 6-Ohda-Induced Partial Lesion of the Nigrostriatal System. Int. J. Neurosci. 2005, 115, 99–117. [Google Scholar] [CrossRef]
  112. Banati, R.B.; Daniel, S.E.; Blunt, S.B. Glial Pathology but Absence of Apoptotic Nigral Neurons in Long-Standing Parkinson’s Disease. Mov. Disord. 1998, 13, 221–227. [Google Scholar] [CrossRef] [PubMed]
  113. Tong, J.; Ang, L.-C.; Williams, B.; Furukawa, Y.; Fitzmaurice, P.; Guttman, M.; Boileau, I.; Hornykiewicz, O.; Kish, S.J. Low Levels of Astroglial Markers in Parkinson’s Disease: Relationship to α-Synuclein Accumulation. Neurobiol. Dis. 2015, 82, 243–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Jyothi, H.J.; Vidyadhara, D.J.; Mahadevan, A.; Philip, M.; Parmar, S.K.; Manohari, S.G.; Shankar, S.K.; Raju, T.R.; Alladi, P.A. Aging Causes Morphological Alterations in Astrocytes and Microglia in Human Substantia Nigra Pars Compacta. Neurobiol. Aging 2015, 36, 3321–3333. [Google Scholar] [CrossRef] [PubMed]
  115. Damier, P.; Hirsch, E.C.; Zhang, P.; Agid, Y.; Javoy-Agid, F. Glutathione Peroxidase, Glial Cells and Parkinson’s Disease. Neuroscience 1993, 52, 1–6. [Google Scholar] [CrossRef] [PubMed]
  116. Tang, Y.; Han, L.; Li, S.; Hu, T.; Xu, Z.; Fan, Y.; Liang, X.; Yu, H.; Wu, J.; Wang, J. Plasma GFAP in Parkinson’s Disease with Cognitive Impairment and Its Potential to Predict Conversion to Dementia. NPJ Park. Dis. 2023, 9, 23. [Google Scholar] [CrossRef]
  117. Su, W.; Chen, H.B.; Li, S.H.; Wu, D.Y. Correlational Study of the Serum Levels of the Glial Fibrillary Acidic Protein and Neurofilament Proteins in Parkinson’s Disease Patients. Clin. Neurol. Neurosurg. 2012, 114, 372–375. [Google Scholar] [CrossRef]
  118. Mao, S.; Teng, X.; Li, Z.; Zu, J.; Zhang, T.; Xu, C.; Cui, G. Association of Serum Neurofilament Light Chain and Glial Fibrillary Acidic Protein Levels with Cognitive Decline in Parkinson’s Disease. Brain Res. 2023, 1805, 148271. [Google Scholar] [CrossRef]
  119. Lin, Y.-Q.; Chen, S.-D. RBD: A Red Flag for Cognitive Impairment in Parkinson’s Disease? Sleep Med. 2018, 44, 38–44. [Google Scholar] [CrossRef]
  120. Teng, X.; Mao, S.; Wu, H.; Shao, Q.; Zu, J.; Zhang, W.; Zhou, S.; Zhang, T.; Zhu, J.; Cui, G.; et al. The Relationship between Serum Neurofilament Light Chain and Glial Fibrillary Acidic Protein with the REM Sleep Behavior Disorder Subtype of Parkinson’s Disease. J. Neurochem. 2023, 165, 268–276. [Google Scholar] [CrossRef]
  121. Oeckl, P.; Halbgebauer, S.; Anderl-Straub, S.; Steinacker, P.; Huss, A.M.; Neugebauer, H.; von Arnim, C.A.F.; Diehl-Schmid, J.; Grimmer, T.; Kornhuber, J.; et al. Glial Fibrillary Acidic Protein in Serum Is Increased in Alzheimer’s Disease and Correlates with Cognitive Impairment. J. Alzheimer’s Dis. 2019, 67, 481–488. [Google Scholar] [CrossRef]
  122. Benussi, A.; Cantoni, V.; Rivolta, J.; Archetti, S.; Micheli, A.; Ashton, N.; Zetterberg, H.; Blennow, K.; Borroni, B. Classification Accuracy of Blood-Based and Neurophysiological Markers in the Differential Diagnosis of Alzheimer’s Disease and Frontotemporal Lobar Degeneration. Alzheimers Res. Ther. 2022, 14, 155. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Brain-biomarker changes in bodily fluids of PD patients. Figure legend: The figure shows proteins that change in the brain (nigrostriatal areas) and bodily fluids (plasma, saliva, and urine) of PD patients. NF-L and ChA were found to be abnormally aggregated in the brain and increased in the blood. SP and P11 decreased in the brain (and SP also decreased in sputum), but increased in the blood, while GFAP and NT increased in both the brain and blood. In contrast, VGF decreased in the brain, but also in blood and urine. NF-L: neurofilament light protein; SP: substance P; P11: S100A10 (S100 calcium-binding protein A10); NT: neurotensin; ChA: Chromogranin A; VGF: no acronym; GFAP: glial fibrillary acidic protein. Upward arrow: increase; downward arrow: decrease.
Figure 1. Brain-biomarker changes in bodily fluids of PD patients. Figure legend: The figure shows proteins that change in the brain (nigrostriatal areas) and bodily fluids (plasma, saliva, and urine) of PD patients. NF-L and ChA were found to be abnormally aggregated in the brain and increased in the blood. SP and P11 decreased in the brain (and SP also decreased in sputum), but increased in the blood, while GFAP and NT increased in both the brain and blood. In contrast, VGF decreased in the brain, but also in blood and urine. NF-L: neurofilament light protein; SP: substance P; P11: S100A10 (S100 calcium-binding protein A10); NT: neurotensin; ChA: Chromogranin A; VGF: no acronym; GFAP: glial fibrillary acidic protein. Upward arrow: increase; downward arrow: decrease.
Ijms 24 10932 g001
Table 1. Changes in protein biomarkers in brains and bodily fluids of PD patients.
Table 1. Changes in protein biomarkers in brains and bodily fluids of PD patients.
Protein Brain Body Fluid
MethodChangeSamplePhaseMethodChangeSamplePhase
NF-LIHC [17]*SNAdvanced
Early
SIMOA [21,22,23,24]
ECLIA [25]
SerumAdvanced Prodromic
ECLIA [20]PlasmaAdvanced
SPRIA [44,45,46]SNAdvancedELISA [50]Serum Advanced
RIA [45,47]PutamenAdvancedELISA [49]SputumEarly
Advanced
RIA [48]CaudateAdvanced
RIA [46]GP Advanced
P11qPCR [53]SNAdvancedFlow cytometry [53]Plasma Advanced
NTHPLCRIA [58,59]
SN
Hyp.
AdvancedRIA [60]PlasmaEarly
Advanced
ChAIHC [66,67]*SN
cerebellum
cerebrum
AdvancedELISA [68]SerumEarly Intermediate
Advanced
VGF ELISA
[87,88,89]
CortexAdvancedELISA [87] PlasmaEarly
Advanced
LC-MS/MS [91]Urinewith or without
LRRK2 G2019S mutation
GFAPIHC [101]SNAdvancedSIMOA [102]PlasmaAdvanced
ELISA [103]
[104]
SIMOA [106]
SerumAdvanced
PD = Parkinson’s disease; NFL = neurofilament light chain; IHC = immunochemistry; * = abnormal aggregation; SN = substantia nigra; SIMOA = single-molecule array; ECLIA = electrochemiluminescence immunoassay; SP = substance P; RIA = radioimmunoassay; ELISA = enzyme-linked immunosorbent assay; GP = globus pallidus; P11 = S100A10 (S100 calcium-binding protein A10); qPCR = quantitative polymerase chain reaction; NT = neurotensin; HPLCRIA = high-performance liquid chromatography with radioimmunoassay; ChA = chromogranin A; SgII = secretogranin II; VGF = non-acronym; Ct = C-terminus; LC/MS/MS = liquid chromatography/mass spectrometry/mass spectrometry; LRRK2 = leucine-rich repeat kinase2; G2019S = Glycine2019Serine mutation; GFAP = glial fibrillary acidic protein. Hyp: hypothalamus. ↑ = increased concentration; ↓ = decreased concentration.
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

Cocco, C.; Manai, A.L.; Manca, E.; Noli, B. Brain-Biomarker Changes in Body Fluids of Patients with Parkinson’s Disease. Int. J. Mol. Sci. 2023, 24, 10932. https://doi.org/10.3390/ijms241310932

AMA Style

Cocco C, Manai AL, Manca E, Noli B. Brain-Biomarker Changes in Body Fluids of Patients with Parkinson’s Disease. International Journal of Molecular Sciences. 2023; 24(13):10932. https://doi.org/10.3390/ijms241310932

Chicago/Turabian Style

Cocco, Cristina, Antonio Luigi Manai, Elias Manca, and Barbara Noli. 2023. "Brain-Biomarker Changes in Body Fluids of Patients with Parkinson’s Disease" International Journal of Molecular Sciences 24, no. 13: 10932. https://doi.org/10.3390/ijms241310932

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