1
|
Ramadan ME, Mostafa TM, Ghali AA, El-Afify DR. Randomized controlled trial evaluating synbiotic supplementation as an adjuvant therapy in the treatment of Parkinson's disease. Inflammopharmacology 2025:10.1007/s10787-025-01752-8. [PMID: 40434674 DOI: 10.1007/s10787-025-01752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/04/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND AND AIM Neuroinflammatory mechanisms have been closely related to the microbiota-gut-brain axis and could lead to degeneration of dopaminergic neurons with subsequent development and progression of Parkinson's disease (PD). Targeting this pathway for the treatment of PD has sparked a lot of interest. Hence, this study investigates the therapeutic potential of a synbiotic supplement, in conjunction with L-dopa for the management of PD. METHODS This randomized controlled trial enrolled 66 Parkinson's disease patients, who were randomly assigned to two groups: a control group (n = 33) receiving standard L-dopa/carbidopa (100/25 mg) therapy three times daily for three months, and a synbiotic group (n = 33) receiving the same L-dopa/carbidopa regimen with two sachets of the synbiotic supplement daily for three months. The outcome measures included assessment of Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and serum levels of tumor necrosis factor-alpha (TNF-α), malondialdehyde (MDA), brain-derived neurotrophic factor (BDNF), and α-synuclein (α-Syn). Blood samples were collected from all patients for biomarker analysis in serum. RESULTS Three months after intervention, the synbiotic group demonstrated significantly greater improvement in motor and non-motor symptoms compared to the control group evidenced by the change in the scores of each part of the MDS-UPDRS. Concurrently, the synbiotic group exhibited significantly lower serum levels of pro-inflammatory marker TNF-α and oxidative stress marker MDA, and significantly higher levels of the neuroprotective factor BDNF. CONCLUSION Supplementing with synbiotics exhibits promising neuroprotective and therapeutic effects in the treatment of Parkinson's disease patients. CLINICALTRIALS GOV REGISTRATION NUMBER NCT05576818. Retrospectively registered in October 2022.
Collapse
Affiliation(s)
- Mohamed E Ramadan
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, 31111, Egypt.
| | - Tarek M Mostafa
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, 31111, Egypt
| | - Azza A Ghali
- Neuropsychiatry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia R El-Afify
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, 31111, Egypt
| |
Collapse
|
2
|
Pons ML, Mohaupt P, Vialaret J, Mondesert E, Vignon M, Coppens S, Stéphane M, Lehmann S, Hirtz C. Plasma α-synuclein domain profiles across α-synucleinopathies. Brain Commun 2025; 7:fcaf189. [PMID: 40433114 PMCID: PMC12107063 DOI: 10.1093/braincomms/fcaf189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
The differential diagnosis of α-synucleinopathies, including Parkinson's disease, dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), remains challenging due to overlapping clinical features and the absence of reliable biomarkers. We developed a targeted mass spectrometry assay to profile α-synuclein peptides in plasma from Parkinson's disease (n = 82), DLB (n = 32), MSA (n = 8) and controls (n = 21). We hypothesized that disease-specific truncations or post-translational modifications would alter levels of non-modified α-synuclein peptides across α-synucleinopathies. The assay quantified non-modified peptides derived from the N-terminus and non-amyloid component (NAC) domain, regions implicated in aggregate formation. Although peptide levels were consistent across disease groups, a distinct NAC domain pattern observed in MSA may reflect unique pathological processes. This study presents the first blood-based profiling of α-synuclein peptides in these disorders, offering a basis for further investigation into disease mechanisms. Refinement of the assay to include post-translational modifications could enhance understanding of α-synucleinopathies and support future biomarker development.
Collapse
Affiliation(s)
- Marie-Laure Pons
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
- Shimadzu Corporation, LC-MS & Life Sciences, Duisburg 47269, Germany
| | - Pablo Mohaupt
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Jérôme Vialaret
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Etienne Mondesert
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Margaux Vignon
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Salomé Coppens
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Moreau Stéphane
- Shimadzu Corporation, LC-MS & Life Sciences, Duisburg 47269, Germany
| | - Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| | - Christophe Hirtz
- LBPC-PPC, Université de Montpellier, IRMB CHU de Montpellier, INM INSERM, Montpellier 34295, France
| |
Collapse
|
3
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
4
|
Li R, Zhuang J. Diagnostic values of serum levels of α-synuclein, homocysteine and leucine-rich α2 glycoprotein for predicting cognitive impairment in Parkinson's disease. Acta Neurol Belg 2025; 125:435-444. [PMID: 39738968 DOI: 10.1007/s13760-024-02710-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND α-synuclein, homocysteine (Hcy) and leucine-rich α2-glycoprotein (LRG) have been shown to correlate to Parkinson's disease (PD). However, it remains unclear whether these factors are associated with the occurrence of cognitive impairment (CI) in PD patients. METHODS This study initially enrolled eligible PD patients without cognitive impairment. Blood samples were collected to measure serum levels of α-synuclein, Hcy, and LRG using enzyme-linked immunosorbent assay. After one year of treatment, patients were divided into CI group and non-CI groups based on their Montreal Cognitive Assessment (MoCA) scores. Baseline clinical characteristics and the levels of these three factors were compared between the two groups. Additionally, Receiver Operating Characteristic (ROC) analysis was used to assess the predictive value of these factors for the occurrence of CI in PD patients. RESULTS The study included 102 eligible PD patients without baseline CI, divided into 65 patients in the non-CI group and 37 patients in the CI group after one year, based on the MoCA scores. Serum levels of α-synuclein, Hcy, and LRG in the CI group were 0.42 ± 0.33 ng/mL, 19.85 ± 6.31 µmol/L, and 14.53 ± 5.11 ng/mL respectively, compared to 0.04 ± 0.03 ng/mL, 14.32 ± 5.25 µmol/L, and 11.67 ± 3.10 ng/mL in the non-CI group. Patients in the CI group had MoCA scores below 26, indicating cognitive impairment. ROC analysis revealed that α-synuclein, Hcy, and LRG levels effectively predicted the occurrence of CI in PD patients. CONCLUSIONS Serum levels of α-synuclein, Hcy and LRG were elevated in the CI group compared to the non-CI group, suggesting that these factors may serve as predictors of cognitive impairment in PD patients.
Collapse
Affiliation(s)
- Rongchuan Li
- Department of Neurology, Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian Campus), No.16 Jingguang Road, Luoshan Section, Jinjiang, 362200, Fujian, China
| | - Jinhong Zhuang
- Department of Neurology, Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian Campus), No.16 Jingguang Road, Luoshan Section, Jinjiang, 362200, Fujian, China.
| |
Collapse
|
5
|
Tahanis A, Hashem V, Ondo W. Serum alpha-synuclein in restless legs syndrome. Sleep 2025; 48:zsae217. [PMID: 39283300 DOI: 10.1093/sleep/zsae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/16/2024] [Indexed: 01/14/2025] Open
Abstract
STUDY OBJECTIVES To determine any correlation between serum alpha-synuclein (α-syn) concentrations and restless legs syndrome (RLS), and to explore the impact of intravenous iron supplementation on serum α-syn levels. METHODS We collected clinical data on 113 RLS patients in whom serum α-syn levels were quantified using an ELISA kit and compared to a group of 45 age-matched controls. A subset of nine RLS patients who received intravenous (IV) iron underwent pre- and post-treatment blood sampling to assess α-syn and ferritin response. RESULTS A family history of RLS was reported by 62.8% of patients, and current dopaminergic augmentation was observed in 31.0%. Low serum ferritin levels below 75 μg/L were seen in 39.8%. Serum α-syn levels were found to be significantly decreased in RLS patients (mean: 7.7 ng/mL) compared to controls (mean: 10.7 ng/mL), p < .05. Stratification based on sex, age, and age of onset, did not reveal significant differences in α-syn levels. In nine RLS patients who received IV iron treatment, a linear correlation between fold change in α-syn and ferritin was observed (R: 0.7, p < .05). The temporal relation between serum α-syn and IV iron treatment showed a gradual decline of α-syn and ferritin by time correlation (p = .023, R: -.739). CONCLUSION In our study of 113 RLS participants, serum α-syn levels were decreased in RLS patients compared to healthy controls, and increased in the nine patients who received IV iron treatment in correlation with ferritin. This correlation could suggest a mechanism for reduced dopamine transmission in RLS.
Collapse
Affiliation(s)
- Aboud Tahanis
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Vera Hashem
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
| | - William Ondo
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
- Department of Neurology, Weill Cornell Medical School, New York, NY, USA
| |
Collapse
|
6
|
Maddocks GM, Eisenstein M, Soh HT. Biosensors for Parkinson's Disease: Where Are We Now, and Where Do We Need to Go? ACS Sens 2024; 9:4307-4327. [PMID: 39189973 DOI: 10.1021/acssensors.4c00790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Parkinson's Disease is the second most common neurological disease in the United States, yet there is no cure, no pinpointed cause, and no definitive diagnostic procedure. Parkinson's is typically diagnosed when patients present with motor symptoms such as slowness of movement and tremors. However, none of these are specific to Parkinson's, and a confident diagnosis of Parkinson's is typically only achieved when 60-80% of dopaminergic neurons are no longer functioning, at which point much of the damage to the brain is irreversible. This Perspective details ongoing efforts and accomplishments in biosensor research with the goal of overcoming these issues for Parkinson's diagnosis and care, with a focus on the potential impact of early diagnosis and associated opportunities to pinpoint a cause and a cure. We critically analyze the strengths and shortcomings of current technologies and discuss the ideal characteristics of a diagnostic technology toolbox to guide future research decisions in this space. Finally, we assess what role biosensors can play in facilitating precision medicine for Parkinson's patients.
Collapse
Affiliation(s)
- Grace M Maddocks
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - M Eisenstein
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - H Tom Soh
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
7
|
Krawczuk D, Kulczyńska-Przybik A, Mroczko B. Clinical Application of Blood Biomarkers in Neurodegenerative Diseases-Present and Future Perspectives. Int J Mol Sci 2024; 25:8132. [PMID: 39125699 PMCID: PMC11311320 DOI: 10.3390/ijms25158132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Neurodegenerative diseases are a group of complex diseases characterized by a progressive loss of neurons and degeneration in different areas of the nervous system. They share similar mechanisms, such as neuroinflammation, oxidative stress, and mitochondrial injury, resulting in neuronal loss. One of the biggest challenges in diagnosing neurodegenerative diseases is their heterogeneity. Clinical symptoms are usually present in the advanced stages of the disease, thus it is essential to find optimal biomarkers that would allow early diagnosis. Due to the development of ultrasensitive methods analyzing proteins in other fluids, such as blood, huge progress has been made in the field of biomarkers for neurodegenerative diseases. The application of protein biomarker measurement has significantly influenced not only diagnosis but also prognosis, differentiation, and the development of new therapies, as it enables the recognition of early stages of disease in individuals with preclinical stages or with mild symptoms. Additionally, the introduction of biochemical markers into routine clinical practice may improve diagnosis and allow for a stratification group of people with higher risk, as well as an extension of well-being since a treatment could be started early. In this review, we focus on blood biomarkers, which could be potentially useful in the daily medical practice of selected neurodegenerative diseases.
Collapse
Affiliation(s)
- Daria Krawczuk
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (A.K.-P.)
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (A.K.-P.)
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (A.K.-P.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland
| |
Collapse
|
8
|
Akowuah PK, Owusu E, Totoe D. Tear α-synuclein as a biomarker for Parkinson's disease: A systematic review and meta-analysis. Optom Vis Sci 2024; 101:485-492. [PMID: 39094023 DOI: 10.1097/opx.0000000000002168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Parkinson's disease symptoms mostly manifest after significant and irreversible neuropathology. Hence, there is a need to identify biomarkers that can provide indications of disease before significant neuronal degeneration occurs. OBJECTIVE To estimate the difference in the concentration of α-synuclein protein in tears between individuals with Parkinson's disease and healthy controls. DATA SOURCES PubMed, Scopus, and Web of Science. The last database search was on December 20, 2023. STUDY ELIGIBILITY CRITERIA Primary prospective studies in humans measuring the level of α-synuclein in tears and clinical outcomes reported using mean or median. PARTICIPANTS AND INTERVENTIONS Individuals with Parkinson's disease and healthy controls. STUDY APPRAISAL AND SYNTHESIS METHODS The risk of bias was assessed using the Newcastle-Ottawa Scale. The I2 statistic was used to estimate heterogeneity. The outcome measure was the difference in tear total and oligomeric α-synuclein. Mean difference (MD) was used to assess the outcome. The certainty of evidence was rated following the Grading of Recommendations Assessment and Development and Evaluation (GRADE) system. RESULTS Three hundred twenty-seven Parkinson's disease and 312 healthy control subjects from five studies and 177 Parkinson's disease and 166 healthy control subjects from two studies were included in total α-synuclein levels and oligomeric α-synuclein levels analysis, respectively. Total α-synuclein level was not different between Parkinson's disease and healthy controls (MD = 0.02 ng/mL [95% confidence interval {CI}: 0.00 to 0.05 ng/mL; I2 = 90%; Z = 1.79; p=0.07; number of studies = 5; GRADE rating = very low]). Stratifying the data based on disease duration, total α-synuclein was higher in subjects with Parkinson's disease duration ≥7 years compared with healthy controls (MD = 0.04 ng/mL [95% CI: 0.03 to 0.05 ng/mL; I2 = 0%; Z = 8.24, p<0.00001; number of studies = 2; GRADE rating = low]) but not different between the two groups (MD = -0.12 ng/mL (95% CI: -0.38 to 0.15 ng/mL; I2 = 93%; Z = 0.84, p=0.40; number of studies = 3; GRADE rating = very low]). Oligomeric α-synuclein level was higher in Parkinson's disease compared with controls (MD = 6.50 ng/mL [95% CI: 2.79 to 10.20 ng/mL; I2 = 94%; Z = 3.44; p=0.0006; number of studies = 2; GRADE rating = very low]). LIMITATIONS High heterogeneity between studies. Potential sources of heterogeneity could not be explored due to the limited number of studies. CONCLUSIONS AND IMPLICATIONS OF KEY FINDINGS Tear α-synuclein has the potential to be a noninvasive biomarker for Parkinson's disease. Studies are, however, needed to increase certainty in the biomarker and establish how the protein's changes in tears correlate with Parkinson's disease progression and severity.
Collapse
Affiliation(s)
| | - Ebenezer Owusu
- College of Optometry, University of Houston, Houston, Texas
| | | |
Collapse
|
9
|
Massey RS, Johri S, Chan D, Holahan MR, Prakash R. Comparison of Aptamer and Antibody Bioreceptors in the OEGFET Biosensor Platform for Detecting α-Synuclein, a Parkinson's Biomarker. IEEE SENSORS LETTERS 2024; 8:1-4. [DOI: 10.1109/lsens.2024.3413575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Roslyn S. Massey
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Srishti Johri
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Dennis Chan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Matthew R. Holahan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
10
|
Cristiani CM, Scaramuzzino L, Quattrone A, Parrotta EI, Cuda G, Quattrone A. Serum Oligomeric α-Synuclein and p-tau181 in Progressive Supranuclear Palsy and Parkinson's Disease. Int J Mol Sci 2024; 25:6882. [PMID: 38999992 PMCID: PMC11241320 DOI: 10.3390/ijms25136882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Clinical differentiation of progressive supranuclear palsy (PSP) from Parkinson's disease (PD) is challenging due to overlapping phenotypes and the late onset of specific atypical signs. Therefore, easily assessable diagnostic biomarkers are highly needed. Since PD is a synucleopathy while PSP is a tauopathy, here, we investigated the clinical usefulness of serum oligomeric-α-synuclein (o-α-synuclein) and 181Thr-phosphorylated tau (p-tau181), which are considered as the most important pathological protein forms in distinguishing between these two parkinsonisms. We assessed serum o-α-synuclein and p-tau181 by ELISA and SIMOA, respectively, in 27 PSP patients, 43 PD patients, and 39 healthy controls (HC). Moreover, we evaluated the correlation between serum biomarkers and biological and clinical features of these subjects. We did not find any difference in serum concentrations of p-tau181 and o-α-synuclein nor in the o-α-synuclein/p-tau181 ratio between groups. However, we observed that serum p-tau181 positively correlated with age in HC and PD, while serum o-α-synuclein correlated positively with disease severity in PD and negatively with age in PSP. Finally, the o-α-synuclein/p-tau181 ratio showed a negative correlation with age in PD.
Collapse
Affiliation(s)
- Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Luana Scaramuzzino
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Andrea Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| | - Elvira Immacolata Parrotta
- Institute of Molecular Biology, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Department of Clinical and Experimental Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Aldo Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (C.M.C.)
| |
Collapse
|
11
|
Shu H, Zhang P, Gu L. Alpha-synuclein in peripheral body fluid as a biomarker for Parkinson's disease. Acta Neurol Belg 2024; 124:831-842. [PMID: 38170418 DOI: 10.1007/s13760-023-02452-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE Whether alpha-synuclein in peripheral body fluids can be used for the diagnosis of Parkinson's disease (PD) remains in controversy. This study evaluates diagnostic potential of alpha-synuclein for PD in various peripheral body fluids using a meta-analysis approach. METHODS Studies published before October 2022 were searched in Web of Science and PubMed databases. The results were computed using the STATA 12.0 statistical software. RESULTS In plasma, PD patients exhibited elevated alpha-synuclein levels relative to healthy controls (HCs) [standard mean difference (SMD) = 0.78, 95% confidence interval (CI) = 0.42 to 1.15] with a sensitivity of 0.79 (95% CI: 0.64-0.89) and a specificity of 0.95 (95% CI: 0.90-0.98). Higher plasma alpha-synuclein levels were correlated with longer disease durations, higher Unified Parkinson's Disease Rating Scale motor scores, and higher Hoehn and Yahr stages in PD patients. Plasma neural-derived exosomal alpha-synuclein levels (SMD = 1.82, 95% CI = 0.30 to 3.35), ratio of plasma neural-derived exosomal alpha-synuclein to total alpha-synuclein (SMD = 1.26, 95% CI = 0.19 to 2.33), and erythrocytic alpha-synuclein levels were also increased in PD patients (SMD = 6.57, 95% CI = 3.55 to 9.58). In serum, there was no significant difference in alpha-synuclein levels between PD patients and HCs (SMD = 0.54, 95% CI = - 0.27 to 1.34). In saliva, reduced alpha-synuclein levels were observed in PD patients (SMD = - 0.85, 95% CI = - 1.67 to - 0.04). CONCLUSIONS Alpha-synuclein levels in plasma, plasma neural-derived exosome, erythrocyte, and saliva may serve as potential biomarkers for the diagnosis of PD.
Collapse
Affiliation(s)
- Hao Shu
- Department of Neurology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Pengcheng Zhang
- Institute of Environment and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300041, China
| | - Lihua Gu
- Department of Neurology, Tianjin Huanhu Hospital, No. 6 Jizhao Road, Tianjin, 300350, China.
| |
Collapse
|
12
|
Alnaaim SA, Al-Kuraishy HM, Alexiou A, Papadakis M, Saad HM, Batiha GES. Role of Brain Liver X Receptor in Parkinson's Disease: Hidden Treasure and Emerging Opportunities. Mol Neurobiol 2024; 61:341-357. [PMID: 37606719 PMCID: PMC10791998 DOI: 10.1007/s12035-023-03561-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease due to the degeneration of dopaminergic neurons (DNs) in the substantia nigra (SN). The liver X receptor (LXR) is involved in different neurodegenerative diseases. Therefore, the objective of the present review was to clarify the possible role of LXR in PD neuropathology. LXRs are the most common nuclear receptors of transcription factors that regulate cholesterol metabolism and have pleiotropic effects, including anti-inflammatory effects and reducing intracellular cholesterol accumulation. LXRs are highly expressed in the adult brain and act as endogenous sensors for intracellular cholesterol. LXRs have neuroprotective effects against the development of neuroinflammation in different neurodegenerative diseases by inhibiting the expression of pro-inflammatory cytokines. LXRs play an essential role in mitigating PD neuropathology by reducing the expression of inflammatory signaling pathways, neuroinflammation, oxidative stress, mitochondrial dysfunction, and enhancement of BDNF signaling.In conclusion, LXRs, through regulating brain cholesterol homeostasis, may be effectual in PD. Also, inhibition of node-like receptor pyrin 3 (NLRP3) inflammasome and nuclear factor kappa B (NF-κB) by LXRs could effectively prevent neuroinflammation in PD. Taken together, LXRs play a crucial role in PD neuropathology by inhibiting neuroinflammation and associated degeneration of DNs.
Collapse
Affiliation(s)
- Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, 14132, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
13
|
Zhang YP, Lobanova E, Emin D, Lobanov SV, Kouli A, Williams-Gray CH, Klenerman D. Imaging Protein Aggregates in Parkinson's Disease Serum Using Aptamer-Assisted Single-Molecule Pull-Down. Anal Chem 2023; 95:15254-15263. [PMID: 37782556 PMCID: PMC10585954 DOI: 10.1021/acs.analchem.3c02515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
The formation of soluble α-synuclein (α-syn) and amyloid-β (Aβ) aggregates is associated with the development of Parkinson's disease (PD). Current methods mainly focus on the measurement of the aggregate concentration and are unable to determine their heterogeneous size and shape, which potentially also change during the development of PD due to increased protein aggregation. In this work, we introduce aptamer-assisted single-molecule pull-down (APSiMPull) combined with super-resolution fluorescence imaging of α-syn and Aβ aggregates in human serum from early PD patients and age-matched controls. Our diffraction-limited imaging results indicate that the proportion of α-syn aggregates (α-syn/(α-syn+Aβ)) can be used to distinguish PD and control groups with an area under the curve (AUC) of 0.85. Further, super resolution fluorescence imaging reveals that PD serums have a higher portion of larger and rounder α-syn aggregates than controls. Little difference was observed for Aβ aggregates. Combining these two metrics, we constructed a new biomarker and achieved an AUC of 0.90. The combination of the aggregate number and morphology provides a new approach to early PD diagnosis.
Collapse
Affiliation(s)
- Yu P. Zhang
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- UK
Dementia Research Institute at Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Evgeniia Lobanova
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- UK
Dementia Research Institute at Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Derya Emin
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- UK
Dementia Research Institute at Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Sergey V. Lobanov
- Medical
Research Council Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff CF24 4HQ, United Kingdom
| | - Antonina Kouli
- Department
of Clinical Neurosciences, University of
Cambridge, Cambridge CB2 0PY, United Kingdom
| | | | - David Klenerman
- Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- UK
Dementia Research Institute at Cambridge, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
14
|
Taneva SG, Todinova S, Andreeva T. Morphometric and Nanomechanical Screening of Peripheral Blood Cells with Atomic Force Microscopy for Label-Free Assessment of Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:14296. [PMID: 37762599 PMCID: PMC10531602 DOI: 10.3390/ijms241814296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegenerative disorders (NDDs) are complex, multifactorial disorders with significant social and economic impact in today's society. NDDs are predicted to become the second-most common cause of death in the next few decades due to an increase in life expectancy but also to a lack of early diagnosis and mainly symptomatic treatment. Despite recent advances in diagnostic and therapeutic methods, there are yet no reliable biomarkers identifying the complex pathways contributing to these pathologies. The development of new approaches for early diagnosis and new therapies, together with the identification of non-invasive and more cost-effective diagnostic biomarkers, is one of the main trends in NDD biomedical research. Here we summarize data on peripheral biomarkers, biofluids (cerebrospinal fluid and blood plasma), and peripheral blood cells (platelets (PLTs) and red blood cells (RBCs)), reported so far for the three most common NDDs-Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). PLTs and RBCs, beyond their primary physiological functions, are increasingly recognized as valuable sources of biomarkers for NDDs. Special attention is given to the morphological and nanomechanical signatures of PLTs and RBCs as biophysical markers for the three pathologies. Modifications of the surface nanostructure and morphometric and nanomechanical signatures of PLTs and RBCs from patients with AD, PD, and ALS have been revealed by atomic force microscopy (AFM). AFM is currently experiencing rapid and widespread adoption in biomedicine and clinical medicine, in particular for early diagnostics of various medical conditions. AFM is a unique instrument without an analog, allowing the generation of three-dimensional cell images with extremely high spatial resolution at near-atomic scale, which are complemented by insights into the mechanical properties of cells and subcellular structures. Data demonstrate that AFM can distinguish between the three pathologies and the normal, healthy state. The specific PLT and RBC signatures can serve as biomarkers in combination with the currently used diagnostic tools. We highlight the strong correlation of the morphological and nanomechanical signatures between RBCs and PLTs in PD, ALS, and AD.
Collapse
Affiliation(s)
- Stefka G. Taneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Tonya Andreeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
- Faculty of Life Sciences, Reutlingen University, Alteburgstraße 150, D-72762 Reutlingen, Germany
| |
Collapse
|
15
|
Kawahata I, Sekimori T, Oizumi H, Takeda A, Fukunaga K. Using Fatty Acid-Binding Proteins as Potential Biomarkers to Discriminate between Parkinson's Disease and Dementia with Lewy Bodies: Exploration of a Novel Technique. Int J Mol Sci 2023; 24:13267. [PMID: 37686075 PMCID: PMC10487513 DOI: 10.3390/ijms241713267] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
An increase in the global aging population is leading to an increase in age-related conditions such as dementia and movement disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and dementia with Lewy bodies (DLB). The accurate prediction of risk factors associated with these disorders is crucial for early diagnosis and prevention. Biomarkers play a significant role in diagnosing and monitoring diseases. In neurodegenerative disorders like α-synucleinopathies, specific biomarkers can indicate the presence and progression of disease. We previously demonstrated the pathogenic impact of fatty acid-binding proteins (FABPs) in α-synucleinopathies. Therefore, this study investigated FABPs as potential biomarkers for Lewy body diseases. Plasma FABP levels were measured in patients with AD, PD, DLB, and mild cognitive impairment (MCI) and healthy controls. Plasma FABP3 was increased in all groups, while the levels of FABP5 and FABP7 tended to decrease in the AD group. Additionally, FABP2 levels were elevated in PD. A correlation analysis showed that higher FABP3 levels were associated with decreased cognitive function. The plasma concentrations of Tau, GFAP, NF-L, and UCHL1 correlated with cognitive decline. A scoring method was applied to discriminate between diseases, demonstrating high accuracy in distinguishing MCI vs. CN, AD vs. DLB, PD vs. DLB, and AD vs. PD. The study suggests that FABPs could serve as potential biomarkers for Lewy body diseases and aid in early disease detection and differentiation.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| | - Tomoki Sekimori
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| | - Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai 982-0805, Japan (A.T.)
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai 982-0805, Japan (A.T.)
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan (K.F.)
| |
Collapse
|
16
|
Al-Kuraishy HM, Al-Gareeb AI, Elewa YHA, Zahran MH, Alexiou A, Papadakis M, Batiha GES. Parkinson's Disease Risk and Hyperhomocysteinemia: The Possible Link. Cell Mol Neurobiol 2023; 43:2743-2759. [PMID: 37074484 PMCID: PMC10333143 DOI: 10.1007/s10571-023-01350-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/09/2023] [Indexed: 04/20/2023]
Abstract
Parkinson's disease (PD) is one of the most common degenerative brain disorders caused by the loss of dopaminergic neurons in the substantia nigra (SN). Lewy bodies and -synuclein accumulation in the SN are hallmarks of the neuropathology of PD. Due to lifestyle changes and prolonged L-dopa administration, patients with PD frequently have vitamin deficiencies, especially folate, vitamin B6, and vitamin B12. These disorders augment circulating levels of Homocysteine with the development of hyperhomocysteinemia, which may contribute to the pathogenesis of PD. Therefore, this review aimed to ascertain if hyperhomocysteinemia may play a part in oxidative and inflammatory signaling pathways that contribute to PD development. Hyperhomocysteinemia is implicated in the pathogenesis of neurodegenerative disorders, including PD. Hyperhomocysteinemia triggers the development and progression of PD by different mechanisms, including oxidative stress, mitochondrial dysfunction, apoptosis, and endothelial dysfunction. Particularly, the progression of PD is linked with high inflammatory changes and systemic inflammatory disorders. Hyperhomocysteinemia induces immune activation and oxidative stress. In turn, activated immune response promotes the development and progression of hyperhomocysteinemia. Therefore, hyperhomocysteinemia-induced immunoinflammatory disorders and abnormal immune response may aggravate abnormal immunoinflammatory in PD, leading to more progression of PD severity. Also, inflammatory signaling pathways like nuclear factor kappa B (NF-κB) and nod-like receptor pyrin 3 (NLRP3) inflammasome and other signaling pathways are intricate in the pathogenesis of PD. In conclusion, hyperhomocysteinemia is involved in the development and progression of PD neuropathology either directly via induction degeneration of dopaminergic neurons or indirectly via activation of inflammatory signaling pathways.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
- Faculty of Veterinary medicine , Hokkaido University, Sapporo, Japan.
| | - Mahmoud Hosny Zahran
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, AlBeheira, 22511, Egypt.
| |
Collapse
|
17
|
Salaramoli S, Joshaghani HR, Hashemy SI. Salivary Biomarkers: Noninvasive Ways for Diagnosis of Parkinson's Disease. Neurol Res Int 2023; 2023:3555418. [PMID: 37434876 PMCID: PMC10332915 DOI: 10.1155/2023/3555418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
Finding reliable biomarkers has a crucial role in Parkinson's disease (PD) assessments. Saliva is a bodily fluid, which might be used as a source of biomarkers for PD. Our article has reviewed several publications on salivary proteins in PD patients and their potential as biomarkers. We find out that α-Syn's proportion in oligomeric form is higher in PD patients' saliva, which is potent to use as a biomarker for PD. The salivary concentration of DJ-1 and alpha-amylase is lower in PD patients. Also, substance P level is more moderate in PD patients. Although salivary flow rate is decreased in PD patients, high levels of heme oxygenase and acetylcholinesterase might be used as noninvasive biomarkers. Salivary miRNAs (miR-153, miR-223, miR-874, and miR-145-3p) are novel diagnostic biomarkers that should be given more attention.
Collapse
Affiliation(s)
- Sanaz Salaramoli
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Joshaghani
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Alrouji M, Al‐kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Jabir MS, Saad HM, Batiha GE. NF-κB/NLRP3 inflammasome axis and risk of Parkinson's disease in Type 2 diabetes mellitus: A narrative review and new perspective. J Cell Mol Med 2023; 27:1775-1789. [PMID: 37210624 PMCID: PMC10315781 DOI: 10.1111/jcmm.17784] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). Genetic predisposition and immune dysfunction are involved in the pathogenesis of PD. Notably, peripheral inflammatory disorders and neuroinflammation are associated with PD neuropathology. Type 2 diabetes mellitus (T2DM) is associated with inflammatory disorders due to hyperglycaemia-induced oxidative stress and the release of pro-inflammatory cytokines. Particularly, insulin resistance (IR) in T2DM promotes the degeneration of dopaminergic neurons in the substantia nigra (SN). Thus, T2DM-induced inflammatory disorders predispose to the development and progression of PD, and their targeting may reduce PD risk in T2DM. Therefore, this narrative review aims to find the potential link between T2DM and PD by investigating the role of inflammatory signalling pathways, mainly the nuclear factor kappa B (NF-κB) and the nod-like receptor pyrin 3 (NLRP3) inflammasome. NF-κB is implicated in the pathogenesis of T2DM, and activation of NF-κB with induction of neuronal apoptosis was also confirmed in PD patients. Systemic activation of NLRP3 inflammasome promotes the accumulation of α-synuclein and degeneration of dopaminergic neurons in the SN. Increasing α-synuclein in PD patients enhances NLRP3 inflammasome activation and the release of interleukin (IL)-1β followed by the development of systemic inflammation and neuroinflammation. In conclusion, activation of the NF-κB/NLRP3 inflammasome axis in T2DM patients could be the causal pathway in the development of PD. The inflammatory mechanisms triggered by activated NLRP3 inflammasome lead to pancreatic β-cell dysfunction and the development of T2DM. Therefore, attenuation of inflammatory changes by inhibiting the NF-κB/NLRP3 inflammasome axis in the early T2DM may reduce future PD risk.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesShaqra UniversityShaqraSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Majid S. Jabir
- Applied Science DepartmentUniversity of TechnologyBaghdadIraq
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
19
|
Brembati V, Faustini G, Longhena F, Bellucci A. Alpha synuclein post translational modifications: potential targets for Parkinson's disease therapy? Front Mol Neurosci 2023; 16:1197853. [PMID: 37305556 PMCID: PMC10248004 DOI: 10.3389/fnmol.2023.1197853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative disorder with motor symptoms. The neuropathological alterations characterizing the brain of patients with PD include the loss of dopaminergic neurons of the nigrostriatal system and the presence of Lewy bodies (LB), intraneuronal inclusions that are mainly composed of alpha-synuclein (α-Syn) fibrils. The accumulation of α-Syn in insoluble aggregates is a main neuropathological feature in PD and in other neurodegenerative diseases, including LB dementia (LBD) and multiple system atrophy (MSA), which are therefore defined as synucleinopathies. Compelling evidence supports that α-Syn post translational modifications (PTMs) such as phosphorylation, nitration, acetylation, O-GlcNAcylation, glycation, SUMOylation, ubiquitination and C-terminal cleavage, play important roles in the modulation α-Syn aggregation, solubility, turnover and membrane binding. In particular, PTMs can impact on α-Syn conformational state, thus supporting that their modulation can in turn affect α-Syn aggregation and its ability to seed further soluble α-Syn fibrillation. This review focuses on the importance of α-Syn PTMs in PD pathophysiology but also aims at highlighting their general relevance as possible biomarkers and, more importantly, as innovative therapeutic targets for synucleinopathies. In addition, we call attention to the multiple challenges that we still need to face to enable the development of novel therapeutic approaches modulating α-Syn PTMs.
Collapse
Affiliation(s)
| | | | | | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
20
|
di Cagno A, Buonsenso A, Centorbi M, Manni L, Di Costanzo A, Casazza G, Parisi A, Guerra G, Calcagno G, Iuliano E, Soligo M, Fiorilli G, for The WB-EMS Parkinson’s Group LenaFrancescoModugnoNicolaQuinziFedericoDepartment of Medicine and Health Sciences, University of Molise, Campobasso, Italy; Neuromed Institute IRCCS, IS, Pozzilli, Italy; Department of Clinical and Experimental Medicine, University of Catanzaro “Magna Græcia”, Italy. Whole body-electromyostimulation effects on serum biomarkers, physical performances and fatigue in Parkinson's patients: A randomized controlled trial. Front Aging Neurosci 2023; 15:1086487. [PMID: 36845654 PMCID: PMC9949720 DOI: 10.3389/fnagi.2023.1086487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Background Whole-body electromyostimulation (WB-EMS) was never previously applied to Parkinson's disease (PD) patients. This randomized controlled study aimed to find the most effective and safe WB-EMS training protocol for this population. Methods Twenty-four subjects (age: 72.13 ± 6.20 years), were randomly assigned to three groups: a high-frequency WB-EMS strength training group (HFG) (rectangular stimulation at 85 Hz, 350 μs, 4 s stimulation/4 s rest), a low-frequency WB-EMS aerobic training group (LFG) (rectangular stimulation 7 Hz, 350 μs, with a continuous pulse duration), and an inactive control group (CG). Participants of the two experimental groups underwent 24 controlled WB-EMS training sessions, with a duration of 20 min each, during 12-week intervention. Serum growth factors (BDNF, FGF-21, NGF and proNGF), α-synuclein, physical performance and Parkinson's Disease Fatigue Scale (PFS-16) responses were analyzed to evaluate the pre-post variation and differences among groups. Results Significant interactions of Time*Groups were detected for BDNF (Time*Groups p = 0.024; Time*CG, b = -628, IC95% = -1,082/-174, p = 0.008), FGF-21 (Time*Groups p = 0.009; Time*LFG b = 1,346, IC95% = 423/2268, p = 0.005), and α-synuclein (Time*Groups p = 0.019; Time*LFG b = -1,572, IC95% = -2,952/-192, p = 0.026). Post hoc analyses and comparisons of ΔS (post-pre), performed independently for each group, showed that LFG increased serum BDNF levels (+ 203 pg/ml) and decreased α-synuclein levels (-1,703 pg/ml), while HFG showed the opposite effects (BDNF: -500 pg/ml; α-synuclein: + 1,413 pg/ml). CG showed a significant BDNF reduction over time. Both LFG and HFG showed significant improvements in several physical performance outcomes and the LFG showed better results than HFG. Concerning PFS-16, significant differences over time (b = -0.4, IC95% = -0.8/-0.0, p = 0.046) and among groups (among all groups p < 0.001) were found, and the LFG exhibited better results than the HFG (b = -1.0, IC95% = -1.3/-0.7, p < 0.001), and CG (b = -1.7, IC95% = -2.0/-1.4, p < 0.001) with this last one that worsened over time. Conclusion LFG training was the best choice for improving or maintaining physical performance, fatigue perception and variation in serum biomarkers. Clinical trial registration https://www.clinicaltrials.gov/ct2/show/NCT04878679, identifier NCT04878679.
Collapse
Affiliation(s)
- Alessandra di Cagno
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Andrea Buonsenso
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Marco Centorbi
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology and Cellular Biology and Neurobiology Institute (CNR), National Research Council (CNR), Rome, Italy
| | - Alfonso Di Costanzo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Giusy Casazza
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Attilio Parisi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Giuseppe Calcagno
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Enzo Iuliano
- Faculty of Psychology, eCampus University, Novedrate, Italy,*Correspondence: Enzo Iuliano,
| | - Marzia Soligo
- Institute of Translational Pharmacology and Cellular Biology and Neurobiology Institute (CNR), National Research Council (CNR), Rome, Italy,Marzia Soligo,
| | - Giovanni Fiorilli
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | |
Collapse
|
21
|
Review of Technological Challenges in Personalised Medicine and Early Diagnosis of Neurodegenerative Disorders. Int J Mol Sci 2023; 24:ijms24043321. [PMID: 36834733 PMCID: PMC9968142 DOI: 10.3390/ijms24043321] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodegenerative disorders are characterised by progressive neuron loss in specific brain areas. The most common are Alzheimer's disease and Parkinson's disease; in both cases, diagnosis is based on clinical tests with limited capability to discriminate between similar neurodegenerative disorders and detect the early stages of the disease. It is common that by the time a patient is diagnosed with the disease, the level of neurodegeneration is already severe. Thus, it is critical to find new diagnostic methods that allow earlier and more accurate disease detection. This study reviews the methods available for the clinical diagnosis of neurodegenerative diseases and potentially interesting new technologies. Neuroimaging techniques are the most widely used in clinical practice, and new techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have significantly improved the diagnosis quality. Identifying biomarkers in peripheral samples such as blood or cerebrospinal fluid is a major focus of the current research on neurodegenerative diseases. The discovery of good markers could allow preventive screening to identify early or asymptomatic stages of the neurodegenerative process. These methods, in combination with artificial intelligence, could contribute to the generation of predictive models that will help clinicians in the early diagnosis, stratification, and prognostic assessment of patients, leading to improvements in patient treatment and quality of life.
Collapse
|
22
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Elekhnawy E. SIRT1 pathway in Parkinson's disease: a faraway snapshot but so close. Inflammopharmacology 2023; 31:37-56. [PMID: 36580159 PMCID: PMC9957916 DOI: 10.1007/s10787-022-01125-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
Silent information regulator (SIRT) has distinctive enzymatic activities and physiological functions to control cell-cycle progression, gene expression, and DNA stability by targeting histone and non-histone proteins. SIRT1 enhances synaptic formation and synaptic activity, and therefore, can reduce the progression of various degenerative brain diseases including Parkinson's disease (PD). SIRT1 activity is decreased by aging with a subsequent increased risk for the development of degenerative brain diseases. Inhibition of SIRT1 promotes inflammatory reactions since SIRT1 inhibits transcription of nuclear factor kappa B (NF-κB) which also inhibits SIRT1 activation via activation of microRNA and miR-34a which reduce NAD synthesis. SIRT1 is highly expressed in microglia as well as neurons, and has antioxidant and anti-inflammatory effects. Therefore, this review aimed to find the possible role of SIRT1 in PD neuropathology. SIRT1 has neuroprotective effects; therefore, downregulation of SIRT1 during aging promotes p53 expression and may increase the vulnerability of neuronal cell deaths. PD neuropathology is linked with the sequence of inflammatory changes and the release of pro-inflammatory cytokines due to the activation of inflammatory signaling pathways. In addition, oxidative stress, inflammatory disorders, mitochondrial dysfunction, and apoptosis contribute mutually to PD neuropathology. Thus, SIRT1 and SIRT1 activators play a crucial role in the mitigation of PD neuropathology through the amelioration of oxidative stress, inflammatory disorders, mitochondrial dysfunction, apoptosis, and inflammatory signaling pathways.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| |
Collapse
|
23
|
Machado S, Teixeira D, Monteiro D, Imperatori C, Murillo-Rodriguez E, da Silva Rocha FP, Yamamoto T, Amatriain-Fernández S, Budde H, Carta MG, Caixeta L, de Sá Filho AS. Clinical applications of exercise in Parkinson's disease: what we need to know? Expert Rev Neurother 2022; 22:771-780. [PMID: 36168890 DOI: 10.1080/14737175.2022.2128768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Exploring the potential of exercise in the rehabilitation process of patients with Parkinson's (PD) may be an interesting treatment perspective. Exercise-induced responses derived from neurotrophic elements appear to ameliorate the decline in neurodegeneration. Despite this understanding, the literature needs to be updated. AREAS COVERED Our review focuses on: a) the key mechanisms of exercise on PD, highlighting mainly the responses related to neuroplasticity; b) the effects induced by different traditional types of exercise, also highlighting the effects of complementary therapies related to movement; c) the volume of exercise required to support efficient results are explored in the context of PD. Additionally, the proposition of new clinical application strategies in the context of PD will also be determined. EXPERT OPINION It is suggested that different intensities of aerobic exercise be explored for the treatment of PD. The results associated with high intensity seem promising for performance, physiological and clinical parameters, such as BDNF production and cognition. On the other hand, the diversification of tasks and repetition of motor gestures appear as consistent arguments to exercise prescription. Finally, for future investigations, the neuromodulation strategy in association with aerobic exercise appears as a potential inducer of benefits on gait and cognitive function.
Collapse
Affiliation(s)
- Sergio Machado
- Department of Sports Methods and Techniques, Federal University of Santa Maria, Santa Maria, Brazil.,Physical Activity Neuroscience Laboratory (LABNAF), Neurodiversity Institute, Queimados-RJ, Brazil.,Intercontinental Neuroscience Research Group, Mérida, Mexico
| | - Diogo Teixeira
- Universidade Lusófona, Faculty of Physical Education and Sport, Lisbon, Portugal; Research Center in Sport, Physical Education, and Exercise and Health (CIDEFES), Lisbon, Portugal
| | - Diogo Monteiro
- ESECS, Polytechnic of Leiria, 2411-901 Leiria, Portugal; Research Center in Sport, Health and Human Development (CIDESD), 5000-558, Vila Real, Portugal.,Life Quality Research Centre (CIEQV), Leiria, Portugal
| | - Claudio Imperatori
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Cognitive and Clinical Psychology Laboratory, Department of Human Sciences European University of Rome, Rome, Italy
| | - Eric Murillo-Rodriguez
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mexico
| | | | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Sandra Amatriain-Fernández
- Institute for Systems Medicine (ISM) at the Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany
| | - Henning Budde
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Institute for Systems Medicine (ISM) at the Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany
| | - Mauro Giovanni Carta
- Dipartimento di Sanità Pubblica, Università degli studi di Cagliari, Cagliari, Italy
| | - Leonardo Caixeta
- Neurology and Neuropsychiatry Department of Clinical Medicine, Federal University of Goiás, School of Medicine, Goiânia, Brazil
| | - Alberto Souza de Sá Filho
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mexico.,Department of Physical Education, Paulista University, Goiânia, Brazil
| |
Collapse
|
24
|
Plasma Phospho-Tau-181 as a Diagnostic Aid in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081879. [PMID: 36009425 PMCID: PMC9405617 DOI: 10.3390/biomedicines10081879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers remain the gold standard for fluid-biomarker-based diagnosis of Alzheimer’s disease (AD) during life. Plasma biomarkers avoid lumbar puncture and allow repeated sampling. Changes of plasma phospho-tau-181 in AD are of comparable magnitude and seem to parallel the changes in CSF, may occur in preclinical or predementia stages of the disease, and may differentiate AD from other causes of dementia with adequate accuracy. Plasma phospho-tau-181 may offer a useful alternative to CSF phospho-tau determination, but work still has to be done concerning the optimal method of determination with the highest combination of sensitivity and specificity and cost-effect parameters.
Collapse
|
25
|
Acquasaliente L, Pontarollo G, Radu CM, Peterle D, Artusi I, Pagotto A, Uliana F, Negro A, Simioni P, De Filippis V. Exogenous human α-Synuclein acts in vitro as a mild platelet antiaggregant inhibiting α-thrombin-induced platelet activation. Sci Rep 2022; 12:9880. [PMID: 35701444 PMCID: PMC9198058 DOI: 10.1038/s41598-022-12886-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
α-Synuclein (αSyn) is a small disordered protein, highly conserved in vertebrates and involved in the pathogenesis of Parkinson’s disease (PD). Indeed, αSyn amyloid aggregates are present in the brain of patients with PD. Although the pathogenic role of αSyn is widely accepted, the physiological function of this protein remains elusive. Beyond the central nervous system, αSyn is expressed in hematopoietic tissue and blood, where platelets are a major cellular host of αSyn. Platelets play a key role in hemostasis and are potently activated by thrombin (αT) through the cleavage of protease-activated receptors. Furthermore, both αT and αSyn could be found in the same spatial environment, i.e. the platelet membrane, as αT binds to and activates platelets that can release αSyn from α-granules and microvesicles. Here, we investigated the possibility that exogenous αSyn could interfere with platelet activation induced by different agonists in vitro. Data obtained from distinct experimental techniques (i.e. multiple electrode aggregometry, rotational thromboelastometry, immunofluorescence microscopy, surface plasmon resonance, and steady-state fluorescence spectroscopy) on whole blood and platelet-rich plasma indicate that exogenous αSyn has mild platelet antiaggregating properties in vitro, acting as a negative regulator of αT-mediated platelet activation by preferentially inhibiting P-selectin expression on platelet surface. We have also shown that both exogenous and endogenous (i.e. cytoplasmic) αSyn preferentially bind to the outer surface of activated platelets. Starting from these findings, a coherent model of the antiplatelet function of αSyn is proposed.
Collapse
Affiliation(s)
- Laura Acquasaliente
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Giulia Pontarollo
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Center for Thrombosis and Hemostasis (CTH) University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Claudia Maria Radu
- Department of Women's & Children's Health, University of Padua, Padua, Italy.,Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy
| | - Daniele Peterle
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Ave. 02115, Boston, MA, USA
| | - Ilaria Artusi
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Anna Pagotto
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Federico Uliana
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Institute of Molecular Systems Biology, ETH Zurich, 8093, Zurich, Switzerland
| | - Alessandro Negro
- Department of Biomedical Sciences, University of Padua, viale G. Colombo 3, 35100, Padua, Italy.
| | - Paolo Simioni
- Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy.
| | - Vincenzo De Filippis
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy. .,Biotechnology Center, CRIBI, University of Padua, Padua, Italy.
| |
Collapse
|
26
|
Cressatti M, Schipper HM. Dysregulation of a Heme Oxygenase-Synuclein Axis in Parkinson Disease. NEUROSCI 2022; 3:284-299. [PMID: 39483365 PMCID: PMC11523740 DOI: 10.3390/neurosci3020020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2024] Open
Abstract
α-Synuclein is a key driver of the pathogenesis of Parkinson disease (PD). Heme oxygenase-1 (HO-1), a stress protein that catalyzes the conversion of heme to biliverdin, carbon monoxide and free ferrous iron, is elevated in PD-affected neural tissues and promotes iron deposition and mitochondrial dysfunction in models of the disease, pathways also impacted by α-synuclein. Elevated expression of human HO-1 in astrocytes of GFAP.HMOX1 transgenic mice between 8.5 and 19 months of age elicits a parkinsonian phenotype characterized by nigrostriatal hypodopaminergia, locomotor incoordination and overproduction of neurotoxic native S129-phospho-α-synuclein. Two microRNAs (miRNA) known to regulate α-synuclein, miR-153 and miR-223, are significantly decreased in the basal ganglia of GFAP.HMOX1 mice. Serum concentrations of both miRNAs progressively decline in wild-type (WT) and GFAP.HMOX1 mice between 11 and 18 months of age. Moreover, circulating levels of miR-153 and miR-223 are significantly lower, and erythrocyte α-synuclein concentrations are increased, in GFAP.HMOX1 mice relative to WT values. MiR-153 and miR-223 are similarly decreased in the saliva of PD patients compared to healthy controls. Upregulation of glial HO-1 may promote parkinsonism by suppressing miR-153 and miR-223, which, in turn, enhance production of neurotoxic α-synuclein. The aim of the current review is to explore the link between HO-1, α-synuclein and PD, evaluating evidence derived from our laboratory and others. HO-1, miR-153 and miR-223 and α-synuclein may serve as potential biomarkers and targets for disease-modifying therapy in idiopathic PD.
Collapse
Affiliation(s)
- Marisa Cressatti
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| | - Hyman M Schipper
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| |
Collapse
|
27
|
Zubelzu M, Morera-Herreras T, Irastorza G, Gómez-Esteban JC, Murueta-Goyena A. Plasma and serum alpha-synuclein as a biomarker in Parkinson's disease: A meta-analysis. Parkinsonism Relat Disord 2022; 99:107-115. [PMID: 35717321 DOI: 10.1016/j.parkreldis.2022.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Reliable biomarkers for Parkinson's disease (PD) diagnosis are urgently needed. Alpha-synuclein (α-syn) and its proteoforms play a key role in PD pathology but in vivo measurements have raised conflicting results, and whether α-syn in blood could distinguish PD patients from healthy controls is still controversial. METHODS A systematic literature search yielded 35 eligible studies for meta-analysis reporting the concentration of total, oligomeric or phosphorylated α-syn in plasma and/or serum of PD patients and healthy controls. Standardized mean differences (SMD) were pooled using multivariate/multilevel linear mixed-effects models. Meta-regression analyses were conducted to investigate possible modifiers. RESULTS A meta-analysis of 32 articles involving 2683 PD patients and 1838 controls showed a significant overall effect of PD on total α-syn levels (SMD = 0.85, p = 0.004). Meta-regression showed that increased SMD of total α-syn in PD was significantly associated with lower age, shorter disease duration, mild motor impairment, and Immunomagnetic Reduction assay for protein quantification. In contrast, no significant differences were observed for oligomeric or phosphorylated α-syn between PD and controls but increased oligomeric α-syn was significantly associated with shorter disease duration. The heterogeneity among studies was high (>98%). CONCLUSIONS These findings suggest that increased total plasma/serum α-syn levels in PD primarily occur in early phases of the disease. The evidence obtained from a small number of studies measuring plasma/serum concentrations of oligomeric and phosphorylated species of α-syn shows no difference. The clinical applicability of measuring plasma or serum α-syn species for differentiating PD from healthy control warrants further studies with better clinical profiling of PD patients.
Collapse
Affiliation(s)
- Maider Zubelzu
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| | - Gorka Irastorza
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Juan Carlos Gómez-Esteban
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Department of Neurology, Cruces University Hospital, Osakidetza, Barakaldo, Bizkaia, Spain
| | - Ane Murueta-Goyena
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
28
|
Gottschalk G, Peterson D, Knox K, Maynard M, Whelan RJ, Roy A. Elevated ATG13 in serum of patients with ME/CFS stimulates oxidative stress response in microglial cells via activation of receptor for advanced glycation end products (RAGE). Mol Cell Neurosci 2022; 120:103731. [PMID: 35487443 DOI: 10.1016/j.mcn.2022.103731] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 10/18/2022] Open
Abstract
Myalgic Encephalomyelitis, also known as Chronic Fatigue Syndrome (ME/CFS), is a multisystem illness characterized by extreme muscle fatigue associated with pain, neurocognitive impairment, and chronic inflammation. Despite intense investigation, the molecular mechanism of this disease is still unknown. Here we demonstrate that autophagy-related protein ATG13 is strongly upregulated in the serum of ME/CFS patients, indicative of impairment in the metabolic events of autophagy. A Thioflavin T-based protein aggregation assay, array screening for autophagy-related factors, densitometric analyses, and confirmation with ELISA revealed that the level of ATG13 was strongly elevated in serum samples of ME/CFS patients compared to age-matched controls. Moreover, our microglia-based oxidative stress response experiments indicated that serum samples of ME/CFS patients evoke the production of reactive oxygen species (ROS) and nitric oxide in human HMC3 microglial cells, whereas neutralization of ATG13 strongly diminishes the production of ROS and NO, suggesting that ATG13 plays a role in the observed stress response in microglial cells. Finally, an in vitro ligand binding assay provided evidence that ATG13 employs the Receptor for Advanced Glycation End-products (RAGE) to stimulate ROS in microglial cells. Collectively, our results suggest that an impairment of autophagy following the release of ATG13 into serum could be a pathological signal in ME/CFS.
Collapse
Affiliation(s)
- Gunnar Gottschalk
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, United States of America; Simmaron R&D lab, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI 53226, United States of America
| | - Daniel Peterson
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, United States of America
| | - Konstance Knox
- Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186., United States of America
| | - Marco Maynard
- Simmaron R&D lab, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI 53226, United States of America
| | - Ryan J Whelan
- Simmaron R&D lab, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI 53226, United States of America
| | - Avik Roy
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, United States of America; Simmaron R&D lab, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI 53226, United States of America.
| |
Collapse
|
29
|
Syafrita Y, Istarini A, Busra M, Indra S, Susanti R. Relationship between Plasma Level of Beta-amyloid, Alpha-synuclein, and Tau Protein with Cognitive Impairment in Parkinson Disease. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Most people with Parkinson’s disease will develop dementia along with their illness development. There are several overlapping brain pathological features in patients with Parkinson's and Alzheimer's disease. These features are related with beta-amyloid findings, alpha-synuclein and tau protein.
Aim: This study was designed to determine the relationship between beta-amyloid, alpha-synuclein and tau protein plasma level with cognitive impairment in Parkinson’s disease.
Materials and Methods: This was an observational with case-control design study. Total of 62 patients with Parkinson’s disease and 20 healthy controls were included in this study. Parkinson’s disease group was divided into 2 subgroups, patient with and without cognitive impairment based on Montreal Cognitive Assessment Indonesian version (MoCA-Ina) score. The plasma levels of beta-amyloid, alpha-synuclein, and tau protein were measured by using enzyme-linked immunoassay technique. Student’s t-test was used to analyze normally distributed data of plasma level differences between groups (Parkinson’s disease group; control group) and subgroups (Parkinson disease with and without cognitive impairment). If the data was not normally distributed, we used Mann-Whitney test. The level of significancy was <0.05 (p value <0.05).
Results: The result demonstrated significant differences in beta-amyloid, alpha-synuclein, and tau protein plasma level between Parkinson’s disease and control group (p<0.05). We also found significant differences of beta-amyloid plasma level between Parkinson’s with and without cognitive impairment subgroups (p<0.05), but none in other parameters (p>0.05).
Conclusion: Low plasma levels of beta-amyloid 42 (Aβ42) are associated with cognitive impairment in patients with Parkinson’s disease.
Collapse
|
30
|
Murueta-Goyena A, Cipriani R, Carmona-Abellán M, Acera M, Ayo N, del Pino R, Tijero B, Fernández T, Gabilondo I, Zallo F, Matute C, Sánchez-Pernaute R, Khurana V, Cavaliere F, Capetillo-Zarate E, Gómez-Esteban JC. Characterization of molecular biomarkers in cerebrospinal fluid and serum of E46K-SNCA mutation carriers. Parkinsonism Relat Disord 2022; 96:29-35. [DOI: 10.1016/j.parkreldis.2022.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
|
31
|
Schumacher-Schuh A, Bieger A, Borelli WV, Portley MK, Awad PS, Bandres-Ciga S. Advances in Proteomic and Metabolomic Profiling of Neurodegenerative Diseases. Front Neurol 2022; 12:792227. [PMID: 35173667 PMCID: PMC8841717 DOI: 10.3389/fneur.2021.792227] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Proteomics and metabolomics are two emerging fields that hold promise to shine light on the molecular mechanisms causing neurodegenerative diseases. Research in this area may reveal and quantify specific metabolites and proteins that can be targeted by therapeutic interventions intended at halting or reversing the neurodegenerative process. This review aims at providing a general overview on the current status of proteomic and metabolomic profiling in neurodegenerative diseases. We focus on the most common neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. We discuss the relevance of state-of-the-art metabolomics and proteomics approaches and their potential for biomarker discovery. We critically review advancements made so far, highlighting how metabolomics and proteomics may have a significant impact in future therapeutic and biomarker development. Finally, we further outline technologies used so far as well as challenges and limitations, placing the current information in a future-facing context.
Collapse
Affiliation(s)
- Artur Schumacher-Schuh
- Departamento de Farmacologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Andrei Bieger
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Wyllians V. Borelli
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Makayla K. Portley
- Neurodegenerative Disorders Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Paula Saffie Awad
- Movement Disorders Clinic, Centro de Trastornos de Movimiento (CETRAM), Santiago, Chile
| | - Sara Bandres-Ciga
- Neurodegenerative Disorders Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Sara Bandres-Ciga
| |
Collapse
|
32
|
Chan DKY, Braidy N, Chen RF, Xu YH, Bentley S, Lubomski M, Davis RL, Chen J, Sue CM, Mellick GD. Strong Predictive Algorithm of Pathogenesis-Based Biomarkers Improves Parkinson's Disease Diagnosis. Mol Neurobiol 2022; 59:1476-1485. [PMID: 34993845 DOI: 10.1007/s12035-021-02604-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/15/2021] [Indexed: 10/19/2022]
Abstract
Easily accessible and accurate biomarkers can aid Parkinson's disease diagnosis. We investigated whether combining plasma levels of α-synuclein, anti-α-synuclein, and/or their ratios to amyloid beta-40 correlated with clinical diagnosis. The inclusion of amyloid beta-40 (Aβ40) is novel. Plasma levels of biomarkers were quantified with ELISA. Using receiver operating characteristic (ROC) curve analysis, levels of α-synuclein, anti-α-synuclein, and their ratios with Aβ40 were analyzed in an initial training set of cases and controls. Promising biomarkers were then used to build a diagnostic algorithm. Verification of the results of biomarkers and the algorithm was performed in an independent set. The training set consisted of 50 cases (age 65.2±9.3, range 44-83, female:male=21:29) with 50 age- and gender-matched controls (67.1±10.0, range 45-96 years; female:male=21:29). ROC curve analysis yielded the following area under the curve results: anti-α-synuclein=0.835, α-synuclein=0.738, anti-α-synuclein/Aβ40=0.737, and α-synuclein/Aβ40=0.663. A 2-step diagnostic algorithm was built: either α-synuclein or anti-α-synuclein was ≥2 times the means of controls (step-1), resulting in 74% sensitivity; and adding α-synuclein/Aβ40 or anti-α-synuclein/Aβ40 (step-2) yielded better sensitivity (82%) while using step-2 alone yielded good specificity in controls (98%). The results were verified in an independent sample of 46 cases and 126 controls, with sensitivity reaching 91.3% and specificity 90.5%. The algorithm was equally sensitive in Parkinson's disease of ≤5-year duration with 92.6% correctly identified in the training set and 90% in the verification set. With two independent samples totaling 272 subjects, our study showed that combination of biomarkers of α-synuclein, anti-α-synuclein, and their ratios to Aβ40 showed promising sensitivity and specificity.
Collapse
Affiliation(s)
- Daniel Kam Yin Chan
- University of New South Wales, Kensington, Australia. .,Western Sydney University, Sydney, Australia. .,Bankstown-Lidcombe Hospital, Eldridge Rd, Bankstown, NSW, 2200, Australia.
| | - Nady Braidy
- University of New South Wales, Kensington, Australia
| | - Ren Fen Chen
- Central Sydney Immunology Laboratory, NSW Health Pathology at Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Ying Hua Xu
- University of New South Wales, Kensington, Australia.,Bankstown-Lidcombe Hospital, Eldridge Rd, Bankstown, NSW, 2200, Australia
| | | | - Michal Lubomski
- Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Ryan L Davis
- Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Jack Chen
- University of New South Wales, Kensington, Australia
| | - Carolyn M Sue
- Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | | |
Collapse
|
33
|
Bougea A, Stefanis L, Chrousos G. Stress system and related biomarkers in Parkinson's disease. Adv Clin Chem 2022; 111:177-215. [DOI: 10.1016/bs.acc.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
34
|
Park DG, Kang J, An YS, Chang J, Yoon JH. Association of plasma α-synuclein with cardiac 123I-MIBG scintigraphy in early Parkinson’s disease. Neurosci Lett 2022; 770:136399. [DOI: 10.1016/j.neulet.2021.136399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/06/2021] [Accepted: 12/13/2021] [Indexed: 01/28/2023]
|
35
|
Ostrakhovitch EA, Song ES, Macedo JKA, Gentry MS, Quintero JE, van Horne C, Yamasaki TR. Analysis of circulating metabolites to differentiate Parkinson's disease and essential tremor. Neurosci Lett 2021; 769:136428. [PMID: 34971771 DOI: 10.1016/j.neulet.2021.136428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022]
Abstract
Parkinson disease (PD) and essential tremor (ET) are two common adult-onset tremor disorders in which prevalence increases with age. PD is a neurodegenerative condition with progressive disability. In ET, neurodegeneration is not an established etiology. We sought to determine whether an underlying metabolic pattern may differentiate ET from PD. Circulating metabolites in plasma and cerebrospinal fluid were analyzed using gas chromatography-mass spectroscopy. There were several disrupted pathways in PD compared to ET plasma including glycolysis, tyrosine, phenylalanine, tyrosine biosynthesis, purine and glutathione metabolism. Elevated α-synuclein levels in plasma and CSF distinguished PD from ET. The perturbed metabolic state in PD was associated with imbalance in the pentose phosphate pathway, deficits in energy production, and change in NADPH, NADH and nicotinamide phosphoribosyltransferase levels. This work demonstrates significant metabolic differences in plasma and CSF of PD and ET patients.
Collapse
Affiliation(s)
| | - Eun-Suk Song
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA
| | - Jessica K A Macedo
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Jorge E Quintero
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Craig van Horne
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Tritia R Yamasaki
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA; Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| |
Collapse
|
36
|
Morphometry and Stiffness of Red Blood Cells—Signatures of Neurodegenerative Diseases and Aging. Int J Mol Sci 2021; 23:ijms23010227. [PMID: 35008653 PMCID: PMC8745649 DOI: 10.3390/ijms23010227] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
Human red blood cells (RBCs) are unique cells with the remarkable ability to deform, which is crucial for their oxygen transport function, and which can be significantly altered under pathophysiological conditions. Here we performed ultrastructural analysis of RBCs as a peripheral cell model, looking for specific signatures of the neurodegenerative pathologies (NDDs)—Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Alzheimer’s disease (AD), utilizing atomic force (AFM) and conventional optical (OM) microscopy. We found significant differences in the morphology and stiffness of RBCs isolated from patients with the selected NDDs and those from healthy individuals. Neurodegenerative pathologies’ RBCs are characterized by a reduced abundance of biconcave discoid shape, lower surface roughness and a higher Young’s modulus, compared to healthy cells. Although reduced, the biconcave is still the predominant shape in ALS and AD cells, while the morphology of PD is dominated by crenate cells. The features of RBCs underwent a marked aging-induced transformation, which followed different aging pathways for NDDs and normal healthy states. It was found that the diameter, height and volume of the different cell shape types have different values for NDDs and healthy cells. Common and specific morphological signatures of the NDDs were identified.
Collapse
|
37
|
Tikader B, Maji SK, Kar S. A generic approach to decipher the mechanistic pathway of heterogeneous protein aggregation kinetics. Chem Sci 2021; 12:13530-13545. [PMID: 34777773 PMCID: PMC8528017 DOI: 10.1039/d1sc03190b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/01/2021] [Indexed: 12/31/2022] Open
Abstract
Amyloid formation is a generic property of many protein/polypeptide chains. A broad spectrum of proteins, despite having diversity in the inherent precursor sequence and heterogeneity present in the mechanism of aggregation produces a common cross β-spine structure that is often associated with several human diseases. However, a general modeling framework to interpret amyloid formation remains elusive. Herein, we propose a data-driven mathematical modeling approach that elucidates the most probable interaction network for the aggregation of a group of proteins (α-synuclein, Aβ42, Myb, and TTR proteins) by considering an ensemble set of network models, which include most of the mechanistic complexities and heterogeneities related to amyloidogenesis. The best-fitting model efficiently quantifies various timescales involved in the process of amyloidogenesis and explains the mechanistic basis of the monomer concentration dependency of amyloid-forming kinetics. Moreover, the present model reconciles several mutant studies and inhibitor experiments for the respective proteins, making experimentally feasible non-intuitive predictions, and provides further insights about how to fine-tune the various microscopic events related to amyloid formation kinetics. This might have an application to formulate better therapeutic measures in the future to counter unwanted amyloidogenesis. Importantly, the theoretical method used here is quite general and can be extended for any amyloid-forming protein.
Collapse
Affiliation(s)
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay Powai Mumbai - 400076 India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay Powai Mumbai - 400076 India
| |
Collapse
|
38
|
Cong S, Xiang C, Wang H, Cong S. Diagnostic utility of fluid biomarkers in multiple system atrophy: a systematic review and meta-analysis. J Neurol 2021; 268:2703-2712. [PMID: 32162061 DOI: 10.1007/s00415-020-09781-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Multiple system atrophy (MSA) is an adult onset, fatal neurodegenerative disease. However, no reliable biomarker is currently available to guide clinical diagnosis and help to determine the prognosis. Thus, a comprehensive meta-analysis is warranted to determine effective biomarkers for MSA and provide useful guidance for clinical diagnosis. METHODS A comprehensive literature search was made of the PubMed, Embase, Cochrane and Web of Science databases for relevant clinical trial articles for 1984-2019. Two review authors examined the full-text records, respectively, and determined which studies met the inclusion criteria. We estimated the mean difference, standard deviation and 95% confidence intervals. RESULTS A total of 28 studies and 11 biomarkers were included in our analysis. Several biomarkers were found to be useful to distinguish MSA patients from healthy controls, including the reduction of phosphorylated tau, α-synuclein (α-syn), 42-amino-acid form of Aβ and total tau (t-tau), the elevation of neurofilament light-chain protein (NFL) in cerebrospinal fluid, the elevation of uric acid and reduction of homocysteine and coenzyme Q10 in plasma. Importantly, α-syn, NFL and t-tau could be used to distinguish MSA from Parkinson's disease (PD), indicating that these three biomarkers could be useful biomarkers in MSA diagnosis. CONCLUSION The findings of our meta-analysis demonstrated diagnostic biomarkers for MSA. Moreover, three biomarkers could be used in differential diagnosis of MSA and PD. The results could be helpful for the early diagnosis of MSA and the accuracy of MSA diagnosis.
Collapse
Affiliation(s)
- Shengri Cong
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Chunchen Xiang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Hailong Wang
- Department of Clinical Epidemiology and Evidence-Based Medicine, First Hospital of China Medical University, Shenyang, China
| | - Shuyan Cong
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
39
|
Agostini S, Mancuso R, Costa AS, Citterio LA, Guerini FR, Meloni M, Navarro J, Clerici M. A Possible Role for HSV-1-Specific Humoral Response and PILRA rs1859788 Polymorphism in the Pathogenesis of Parkinson's Disease. Vaccines (Basel) 2021; 9:686. [PMID: 34206597 PMCID: PMC8310311 DOI: 10.3390/vaccines9070686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022] Open
Abstract
The etiology of Parkinson's disease (PD), a progressive nervous system disorder that affects movement, is still unknown; both genetic and environmental factor are believed to be involved in onset of the disease and its development. Herpes simplex virus type 1 (HSV-1), in particular, is suspected to have a role in PD. Paired Immunoglobulin-like type 2 receptor alpha (PILRA) is an inhibitory receptor that down-regulates inflammation and is expressed on innate immune cells. The PILRA rs1859788 polymorphism is protective against Alzheimer's disease, even in relation with HSV-1 antibody titers, but no data are available in PD. We analyzed HSV-1 antibody titers and PILRA rs1859788 in PD (n = 51) and age-and sex-matched healthy controls (HC; n = 73). Results showed that HSV-1, but not cytomegalovirus (CMV) or human herpes virus type 6 (HHV-6) antibody titers were significantly higher in PD compared to HC (p = 0.045). The rs1859788 polymorphism was not differentially distributed between PD and HC, but the minor allele A was more frequently carried by PD (68%) compared to HC (50%) (p = 0.06). Notably, the rs1859788 minor allele A was statically more frequent in male PD (65%) compared to male HC (37%) (p = 0.036). Finally, no relation was found between HSV-1 antibody titers and PILRA genotype. Results herein suggest an involvement of HSV-1 in PD and indicate a possible interaction between PILRA gene polymorphisms and this neuropathology.
Collapse
Affiliation(s)
- Simone Agostini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Roberta Mancuso
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Andrea S. Costa
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Lorenzo A. Citterio
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Franca R. Guerini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Mario Meloni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Jorge Navarro
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy; (R.M.); (A.S.C.); (L.A.C.); (F.R.G.); (M.M.); (J.N.); (M.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| |
Collapse
|
40
|
Can Plasma α-Synuclein Help Us to Differentiate Parkinson's Disease from Essential Tremor? Tremor Other Hyperkinet Mov (N Y) 2021; 11:20. [PMID: 34113478 PMCID: PMC8162285 DOI: 10.5334/tohm.600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background Studies have revealed controversial results regarding the diagnostic accuracy of plasma α-synuclein levels in patients with Parkinson's disease (PD). This study was aimed to analyze the diagnostic accuracy of plasma α-synuclein in PD versus healthy controls and patients with essential tremor (ET). Methods In this cross-sectional study, we included de novo (n = 19) and advanced PD patients [OFF (n = 33), and On (n = 35) states], patients with ET (n = 19), and controls (n = 35). The total plasma α-synuclein levels were determined using an ELISA sandwich method. We performed adjusted multivariate regression analysis to estimate the association of α-synuclein levels with group conditions [controls, ET, and de novo, OFF and ON-PD]. We studied the diagnostic accuracy of plasma α-synuclein using the area under the curve (AUC). Results The plasma α-synuclein levels were higher in controls compared to PD and ET (p < 0.0001), discriminating de novo PD from controls (AUC = 0.74, 95% CI 0.60-0.89), with a trend towards in advanced PD (OFF state) from ET (AUC = 0.69, 95% CI 0.53-0.84). Conclusions This is the first study examining and comparing plasma α-synuclein levels in ET vs. PD and controls. Preliminary findings suggest that plasma α-synuclein levels might help to discriminate de novo and advanced PD from controls and ET.
Collapse
|
41
|
Chung CC, Chan L, Chen JH, Hung YC, Hong CT. Plasma Extracellular Vesicle α-Synuclein Level in Patients with Parkinson's Disease. Biomolecules 2021; 11:biom11050744. [PMID: 34067663 PMCID: PMC8155846 DOI: 10.3390/biom11050744] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The most established pathognomonic protein of Parkinson's disease (PD), α-synuclein, is extensively investigated for disease diagnosis and prognosis; however, investigations into whether the free form of α-synuclein in the blood functions as a PD biomarker have not been fruitful. Extracellular vesicles (EVs) secreted from cells and present in blood transport molecules are novel platforms for biomarker identification. In blood EVs, α-synuclein originates predominantly from the brain without the interference of the blood-brain barrier. The present study investigated the role of plasma EV-borne α-synuclein as a biomarker of PD. METHODS Patients with mild to moderate stages of PD (n = 116) and individuals without PD (n = 46) were recruited to serve as the PD study group and the control group, respectively. Plasma EVs were isolated, and immunomagnetic reduction-based immunoassay was used to assess EV α-synuclein levels. Conventional statistical analysis was performed using SPSS 25.0, and p < 0.05 was considered significant. RESULTS Compared with controls, we observed significantly lower plasma EV α-synuclein levels in the patients with PD (PD: 56.0 ± 3.7 fg/mL vs. control: 74.5 ± 4.3 fg/mL, p = 0.009), and the significance remained after adjustment for age and sex. Plasma EV α-synuclein levels in the patients with PD did not correlate with age, disease duration, Part I and II scores of the Unified Parkinson's Disease Rating Scale (UPDRS), or the Mini-Mental State Examination scores. However, such levels were significantly correlated with UPDRS Part III score, which assesses motor dysfunction. Furthermore, the severity of akinetic-rigidity symptoms, but not tremor, was inversely associated with plasma EV α-synuclein level. CONCLUSION Plasma EV α-synuclein was significantly different between the control and PD group and was associated with akinetic-rigidity symptom severity in patients with PD. This study corroborates the possible diagnostic and subtyping roles of plasma EV α-synuclein in patients with PD, and it further provides a basis for this protein's clinical relevance and feasibility as a PD biomarker.
Collapse
Affiliation(s)
- Chen-Chih Chung
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 11031, Taiwan
| | - Lung Chan
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
| | - Yi-Chieh Hung
- Department of Neurosurgery, Department of Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
- Correspondence: (Y.-C.H.); (C.-T.H.); Tel.: +886-2-224-900-88 (ext. 811) (C.-T.H.)
| | - Chien-Tai Hong
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-C.H.); (C.-T.H.); Tel.: +886-2-224-900-88 (ext. 811) (C.-T.H.)
| |
Collapse
|
42
|
Association of gender and age at onset with glucocerebrosidase associated Parkinson's disease: a systematic review and meta-analysis. Neurol Sci 2021; 42:2261-2271. [PMID: 33837876 DOI: 10.1007/s10072-021-05230-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/06/2021] [Indexed: 12/17/2022]
Abstract
Glucocerebrosidase (GBA) gene has been proved to be a risk factor for the development of Parkinson's disease (PD). However, the gender effect in the prevalence of GBA-associated PD (GBA-PD) is still controversial. And there is no conclusion whether the age at onset (AAO) of PD is different between carriers and non-carriers of GBA. To clarify the association between gender and AAO in GBA-PD, we conducted a systematic review and meta-analysis. PubMed, Web of Science, and Embase were retrieved to obtain potentially related studies. The odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to determine the association between gender and GBA-PD. And the weighted mean difference (WMD) with 95% CIs was employed to assess the difference of AAO between carriers and non-carriers of GBA. A total of twenty-eight studies involving 16,488 PD patients were included in this meta-analysis. The results showed the prevalence of female patients was higher in GBA-PD [OR: 1.19, (95% CI, 1.07-1.32), P = 0.001]. Meanwhile, GBA carriers had younger age at PD onset than GBA non-carriers [WMD: 2.87, (95% CI, 2.48-3.27), P < 0.001]. Results of subgroup analysis showed the prevalence of women in GBA-PD was higher than men in North American and European PD patients, while the gender difference was not significant in other areas around the world, suggesting an ethnic specificity of gender effect for GBA-PD. Our results indicate the higher female prevalence with ethnic specificity and younger AAO of GBA carriers in GBA-PD.
Collapse
|
43
|
Blanco-Palmero VA, Azcárate-Díaz FJ, Ruiz-Ortiz M, Laespada-García MI, Rábano-Suárez P, Méndez-Guerrero A, Aramendi-Ramos M, Eguiburu JL, Pérez-Rivilla A, Marchán-López A, Rubio-Fernández M, Carro E, González de la Aleja J. Serum and CSF alpha-synuclein levels do not change in COVID-19 patients with neurological symptoms. J Neurol 2021; 268:3116-3124. [PMID: 33606070 PMCID: PMC7892700 DOI: 10.1007/s00415-021-10444-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 infection can associate diverse neurological manifestations. Several studies have provided proof to support the theory of neurotropic involvement of SARS-CoV-2. Alpha-synuclein has been described as a native antiviral factor within neurons, and upregulation of this protein can be seen in animals that suffered other neuroinvasive infections. To assess if increased expression of this protein takes place in COVID-19 patients with neurological symptoms, we analyzed serum total alpha-synuclein levels in three groups: seven COVID-19 patients with myoclonus, Parkinsonism and/or encephalopathy; thirteen age- and sex-matched COVID-19 patients without neurological involvement and eight age- and sex-matched healthy controls. We did not find differences among them. In a subset of four patients, the change in serum alpha-synuclein before and after the onset of neurological symptoms was not significant either. Cerebrospinal fluid alpha-synuclein levels were also similar between neurological COVID-19 and healthy controls. Overall, these results cannot support the hypothesis of alpha-synuclein upregulation in humans with neurological symptoms in COVID-19. Further research taking into account a larger group of COVID-19 patients including the whole spectrum of neurological manifestations and disease severity is needed.
Collapse
Affiliation(s)
- V A Blanco-Palmero
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain. .,Group of Neurodegenerative Diseases, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain. .,Networked Biomedical Research Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - F J Azcárate-Díaz
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| | - M Ruiz-Ortiz
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| | - M I Laespada-García
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| | - P Rábano-Suárez
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| | - A Méndez-Guerrero
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| | - M Aramendi-Ramos
- Department of Clinical Analysis, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J L Eguiburu
- Department of Clinical Analysis, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - A Pérez-Rivilla
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - A Marchán-López
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M Rubio-Fernández
- Group of Neurodegenerative Diseases, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Networked Biomedical Research Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - E Carro
- Group of Neurodegenerative Diseases, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Networked Biomedical Research Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - J González de la Aleja
- Department of Neurology, Hospital Universitario 12 de Octubre, Avda. de Córdoba, s/n 28041, Madrid, Spain
| |
Collapse
|
44
|
Lee SYH, Yates NJ, Tye SJ. Inflammatory Mechanisms in Parkinson's Disease: From Pathogenesis to Targeted Therapies. Neuroscientist 2021; 28:485-506. [PMID: 33586516 DOI: 10.1177/1073858421992265] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Inflammation is a critical factor contributing to the progressive neurodegenerative process observed in Parkinson's disease (PD). Microglia, the immune cells of the central nervous system, are activated early in PD pathogenesis and can both trigger and propagate early disease processes via innate and adaptive immune mechanisms such as upregulated immune cells and antibody-mediated inflammation. Downstream cytokines and gene regulators such as microRNA (miRNA) coordinate later disease course and mediate disease progression. Biomarkers signifying the inflammatory and neurodegenerative processes at play within the central nervous system are of increasing interest to clinical teams. To be effective, such biomarkers must achieve the highest sensitivity and specificity for predicting PD risk, confirming diagnosis, or monitoring disease severity. The aim of this review was to summarize the current preclinical and clinical evidence that suggests that inflammatory processes contribute to the initiation and progression of neurodegenerative processes in PD. In this article, we further summarize the data about main inflammatory biomarkers described in PD to date and their potential for regulation as a novel target for disease-modifying pharmacological strategies.
Collapse
Affiliation(s)
- Stellina Y H Lee
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Nathanael J Yates
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Susannah J Tye
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA.,Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
45
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
46
|
Fricova D, Harsanyiova J, Kralova Trancikova A. Alpha-Synuclein in the Gastrointestinal Tract as a Potential Biomarker for Early Detection of Parkinson's Disease. Int J Mol Sci 2020; 21:E8666. [PMID: 33212934 PMCID: PMC7698349 DOI: 10.3390/ijms21228666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
The primary pathogenesis associated with Parkinson's disease (PD) occurs in peripheral tissues several years before the onset of typical motor symptoms. Early and reliable diagnosis of PD could provide new treatment options for PD patients and improve their quality of life. At present, however, diagnosis relies mainly on clinical symptoms, and definitive diagnosis is still based on postmortem pathological confirmation of dopaminergic neuronal degeneration. In addition, the similarity of the clinical, cognitive, and neuropathological features of PD with other neurodegenerative diseases calls for new biomarkers, suitable for differential diagnosis. Alpha-synuclein (α-Syn) is a potential PD biomarker, due to its close connection with the pathogenesis of the disease. Here we summarize the currently available information on the possible use of α-Syn as a biomarker of early stages of PD in gastrointestinal (GI) tissues, highlight its potential to distinguish PD and other neurodegenerative diseases, and suggest alternative methods (primarily developed for other tissue analysis) that could improve α-Syn detection procedures or diagnostic methods in general.
Collapse
Affiliation(s)
- Dominika Fricova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 814 38 Bratislava, Slovakia;
| | - Jana Harsanyiova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University, 814 99 Bratislava, Slovakia;
| | - Alzbeta Kralova Trancikova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, 814 99 Bratislava, Slovakia
| |
Collapse
|
47
|
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders characterized by gradual progressive neuronal loss in the central nervous system. Unfortunately, the pathogenesis of many of these diseases remains unknown. Synucleins are a family of small, highly charged proteins expressed predominantly in neurons. Following their discovery, much has been learned about their structure, function, interaction with other proteins and role in neurodegenerative disease over the last two decades. One of these proteins, α-Synuclein (α-Syn), appears to be involved in many neurodegenerative disorders. These include Parkinson's disease (PD), dementia with Lewy bodies (DLB), Rapid Eye Movement Sleep Behavior Disorder (RBD) and Pure Autonomic Failure (PAF), i.e., collectively termed α-synucleinopathies. This review focuses on α-Syn dysfunction in neurodegeneration and assesses its role in synucleinopathies from a biochemical, genetic and neuroimaging perspective.
Collapse
Affiliation(s)
- Anastasia Bougea
- Neurochemistry Laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Neuroscience Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
48
|
Fu Y, Jiang C, Tofaris GK, Davis JJ. Facile Impedimetric Analysis of Neuronal Exosome Markers in Parkinson's Disease Diagnostics. Anal Chem 2020; 92:13647-13651. [PMID: 32945162 PMCID: PMC7584333 DOI: 10.1021/acs.analchem.0c03092] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
![]()
The
egress of α-synuclein in neuronally derived exosomes
predates the clinical presentation of Parkinson’s disease (PD),
offering a means of developing a predictive or prognostic test. Here,
we report the reagentless impedimetric assay of two internal exosome
markers (α-synuclein and syntenin-1) from neuronal exosomes.
Exosomes were efficiently extracted from patient sera using anti-L1CAM
conjugated zwitterionic polymer-modified magnetic beads prior to lysis
and analyzed by electrochemical impedance spectroscopy. The quantification
of α-synuclein level across 40 clinical samples resolved statistically
significant differences between PD patients and healthy controls (HC).
Collapse
Affiliation(s)
- Ying Fu
- Department of Chemistry, University of Oxford, Oxford OX1 3QZ, United Kingdom
| | - Cheng Jiang
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Jason J Davis
- Department of Chemistry, University of Oxford, Oxford OX1 3QZ, United Kingdom
| |
Collapse
|
49
|
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease variably associated with motor, nonmotor, and autonomic symptoms, resulting from putaminal and cerebellar degeneration and associated with glial cytoplasmic inclusions enriched with α-synuclein in oligodendrocytes and neurons. Although symptomatic treatment of MSA can provide significant improvements in quality of life, the benefit is often partial, limited by adverse effects, and fails to treat the underlying cause. Consistent with the multisystem nature of the disease and evidence that motor symptoms, autonomic failure, and depression drive patient assessments of quality of life, treatment is best achieved through a coordinated multidisciplinary approach driven by the patient's priorities and goals of care. Research into disease-modifying therapies is ongoing with a particular focus on synuclein-targeted therapies among others. This review focuses on both current management and emerging therapies for this devastating disease.
Collapse
Affiliation(s)
- Matthew R. Burns
- Norman Fixel Institute for Neurological Diseases at UFHealth, Movement Disorders Division, Department of Neurology, University of Florida, 3009 SW Williston Rd, Gainesville, FL 32608 USA
| | - Nikolaus R. McFarland
- Norman Fixel Institute for Neurological Diseases at UFHealth, Movement Disorders Division, Department of Neurology, University of Florida, 3009 SW Williston Rd, Gainesville, FL 32608 USA
| |
Collapse
|
50
|
Alsiary RA, Alghrably M, Saoudi A, Al-Ghamdi S, Jaremko L, Jaremko M, Emwas AH. Using NMR spectroscopy to investigate the role played by copper in prion diseases. Neurol Sci 2020; 41:2389-2406. [PMID: 32328835 PMCID: PMC7419355 DOI: 10.1007/s10072-020-04321-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/29/2020] [Indexed: 12/31/2022]
Abstract
Prion diseases are a group of rare neurodegenerative disorders that develop as a result of the conformational conversion of normal prion protein (PrPC) to the disease-associated isoform (PrPSc). The mechanism that actually causes disease remains unclear. However, the mechanism underlying the conformational transformation of prion protein is partially understood-in particular, there is strong evidence that copper ions play a significant functional role in prion proteins and in their conformational conversion. Various models of the interaction of copper ions with prion proteins have been proposed for the Cu (II)-binding, cell-surface glycoprotein known as prion protein (PrP). Changes in the concentration of copper ions in the brain have been associated with prion diseases and there is strong evidence that copper plays a significant functional role in the conformational conversion of PrP. Nevertheless, because copper ions have been shown to have both a positive and negative effect on prion disease onset, the role played by Cu (II) ions in these diseases remains a topic of debate. Because of the unique properties of paramagnetic Cu (II) ions in the magnetic field, their interactions with PrP can be tracked even at single atom resolution using nuclear magnetic resonance (NMR) spectroscopy. Various NMR approaches have been utilized to study the kinetic, thermodynamic, and structural properties of Cu (II)-PrP interactions. Here, we highlight the different models of copper interactions with PrP with particular focus on studies that use NMR spectroscopy to investigate the role played by copper ions in prion diseases.
Collapse
Affiliation(s)
- Rawiah A. Alsiary
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Mawadda Alghrably
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Abdelhamid Saoudi
- Oncology, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia. King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Suliman Al-Ghamdi
- Oncology, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia. King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Lukasz Jaremko
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Mariusz Jaremko
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Abdul-Hamid Emwas
- Imaging and Characterization Core Lab, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| |
Collapse
|