1
|
Chen YN, Chen YR, Yang BH, Lee PH, Kuo LT, Li CH. Intranasal administration of tissue inhibitor of metalloproteinase 1 extracted from stem cells from human exfoliated deciduous teeth-conditioned medium improves neurological outcomes and alpha-synuclein elimination in Parkinson's disease mice. J Dent Sci 2025; 20:1043-1051. [PMID: 40224104 PMCID: PMC11993047 DOI: 10.1016/j.jds.2024.11.029] [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: 07/10/2024] [Revised: 11/26/2024] [Indexed: 04/15/2025] Open
Abstract
Background/purpose Parkinson's disease (PD) remains a challenging neurodegenerative disorder, requiring the exploration of innovative therapeutic strategies. This study assessed the neuroprotective effects of stem cells from human exfoliated deciduous teeth-conditioned medium (SHED-CM) in a rotenone-induced mouse model of PD. Materials and methods The SHED-CM, fractionated and purified via Fast Protein Liquid Chromatography (FPLC) into "yPD01", was administered intranasally to evaluate its impact on motor deficits, olfactory functions, and protein expressions in affected brain regions. Results Upon intranasal delivery, yPD01 produced significant improvements to motor deficits and restoration of protein expressions associated with PD pathology, particularly in the olfactory bulb and substantial nigra. Intriguingly, the intranasal route exhibited efficacy akin to intravenous administration, highlighting its potential as a minimally invasive yet equally effective therapeutic approach. Further investigation indicated key components within yPD01, denoted as peaks P1 and P5, showcasing pivotal therapeutic effects. These components were linked to the interruption of alpha-synuclein aggregation and its clearance within specific brain cells affected by PD. Conclusion Intranasally administered yPD01 shows neuroprotective potential for mitigating neurodegenerative symptoms, particularly in PD. The results provide valuable insights into potential mechanisms underlying the neuroprotective effects of SHED-CM and their implications for future therapeutic interventions in PD and related conditions.
Collapse
Affiliation(s)
- Yi-No Chen
- Non-invasive Cancer Therapy Research Institute - Taiwan, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yong-Ren Chen
- Non-invasive Cancer Therapy Research Institute - Taiwan, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Bing-Heng Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan
- Trace Element Research Center, Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan
| | - Po-Hui Lee
- Non-invasive Cancer Therapy Research Institute - Taiwan, Taipei, Taiwan
| | - Lu-Ting Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Hsing Li
- School of Dentistry and Graduate Institute of Dental Science, National Defense Medical Center, Taipei, Taiwan
- Division of Orthodontics and Pediatric Dentistry, Department of Dentistry, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Zhang G, Ma C, Ma L, Wei D, Wu Y, Li Y, Xu Z, Liu Y, Cai Y, Yu EY, Zhu Y, Zhang H. High-Efficiency Capture and Proteomic Analysis of Plasma-Derived Extracellular Vesicles through Affinity Purification. Anal Chem 2025; 97:4889-4897. [PMID: 39908429 DOI: 10.1021/acs.analchem.4c04269] [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: 02/07/2025]
Abstract
Plasma-derived extracellular vesicles (EVs) are promising sources of biomarkers. It is still a challenge to isolate EVs from a small amount of human plasma for downstream proteomic analysis. The isolation process is hindered by contamination with high-abundance blood proteins and lipoprotein particles, which adversely impact proteomic analyses. Moreover, although EV immune-isolation via magnetic beads often integrates with flow sorting and Western blotting (WB), it lacks compatibility with nanoparticle tracking analysis (NTA) and proteomic analysis. To address these issues, we have developed a functional affinity magnetic bead, EVlent (Extracellular Vesicles isoLated Efficiently, Naturally, and Totally), enabling the rapid and efficient isolation of EVs from plasma. By optimizing the quantities of magnetic beads and plasma used, we characterized the isolated EVs through WB, NTA, and transmission electron microscopy (TEM), showing the successful isolation of EVs from plasma. Proteomic analysis of these EVs identified over 2000 proteins and 15,000 peptides from 100 μL of plasma and nearly 1000 proteins from trace samples as small as 5 μL. Additionally, this isolation method significantly reduced contaminants, including plasma proteins and lipoproteins, compared to ultracentrifugation. Finally, we applied this strategy to plasma samples of healthy individuals and those with Parkinson's disease, identifying four potential biomarkers that provide promising guidance for clinical diagnosis.
Collapse
Affiliation(s)
- Guiyuan Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- EVLiXiR Biotech, Nanjing 210032, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Chengxiao Ma
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Le Ma
- Shanghai JINCE Clinical Laboratories, Shanghai 201101, China
| | - Dong Wei
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yanan Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Ying Li
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhehui Xu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yufeng Liu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yuhan Cai
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Evan Yiwen Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6229ER, The Netherlands
| | - Yefei Zhu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Department of Laboratory Medicine, Jianhu People's Hospital, Yanchen 224700, China
| | - Hao Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- EVLiXiR Biotech, Nanjing 210032, China
| |
Collapse
|
3
|
Thomsen MS, Kostrikov S, Routhe LG, Johnsen KB, Helgudóttir SS, Gudbergsson JM, Andresen TL, Moos T. Remodeling of the brain angioarchitecture in experimental chronic neurodegeneration. Neurobiol Dis 2025; 204:106761. [PMID: 39662534 DOI: 10.1016/j.nbd.2024.106761] [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: 08/28/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Chronic neurodegenerative diseases are characterized by substantial inflammation with putative impairment of the brain vasculature also commonly observed. To address effects of chronic neurodegeneration on the regional vasculature under experimentally controlled circumstances, the glutamate receptor agonist ibotenic acid was injected into striatum of adult rats, which causes excitotoxicity in the substantia nigra pars reticulata (SNpr) due to imbalance between inhibitory inputs from the striatum and excitatory signals from the subthalamic nucleus. Brains were examined at 28 days (short-term neurodegeneration) and 91 days (long-term neurodegeneration) and analyzed for vascular remodeling taking both 2D and 3D approaches, the latter involving confocal microscopy of optically cleared samples combined with machine learning-based image analysis. Crysectioned and microdissected samples were analyzed for protein and gene expression respectively. The resulting neurodegeneration was accompanied by regional tissue loss and inflammation. The 3D analysis of the degenerating SNpr revealed substantial changes of the vasculature with higher density, increased diameter, and number of tortuous vessels already after 28 days, evidently continuing at 91 days. Interestingly, the vascular remodeling changes occurred without changes in the expression of endothelial tight junction proteins, vascular basement membrane proteins, or markers of angiogenesis. We propose that remodeling of the vasculature in neurodegeneration occurs due to regional tissue atrophy, which leaves the vasculature operating but prone to additional pathologies.
Collapse
Affiliation(s)
- Maj Schneider Thomsen
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Serhii Kostrikov
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lisa Greve Routhe
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Kasper Bendix Johnsen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Steinunn Sara Helgudóttir
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Johann Mar Gudbergsson
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Torben Moos
- Neurobiology research and drug delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
4
|
Scholefield M, Church SJ, Philbert S, Xu J, Patassini S, Cooper GJS. Human dementia with Lewy bodies brain shows widespread urea elevations. Parkinsonism Relat Disord 2024; 124:107017. [PMID: 38788611 DOI: 10.1016/j.parkreldis.2024.107017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/25/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
INTRODUCTION Several recent studies have uncovered the presence of widespread urea elevations in multiple neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease dementia (PDD), vascular dementia (VaD), and Huntington's disease (HD). However, it is currently unknown whether dementia with Lewy bodies also shows these alterations in urea. This study aimed to investigate if and where urea is perturbed in the DLB brain. METHODS Tissues from ten brain regions were obtained from 20 diagnosed cases of DLB and 19 controls. Urea concentrations were measured using ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Case-control differences were assessed by nonparametric Mann-Whitney U tests, and s-values, E-values, effect sizes, and risk ratios were determined for each brain region. The results were compared to those previously obtained for AD, PDD, VaD, and HD. RESULTS As with other previously investigated dementia diseases, DLB shows widespread urea elevations, affecting all ten regions investigated in the current study; the degree of these elevations is lower than that seen in AD or PDD, similar to that seen in HD, and higher than that observed in VaD. The highest urea fold-change was observed in the pons and the lowest in the primary visual cortex. CONCLUSION Urea elevations appear to be a shared alterations across at least five neurodegenerative diseases, despite their many differences in clinical and neuropathological presentation. The cause and effects of this perturbation should be the focus of future studies, for its possible contributions to the pathology of these conditions.
Collapse
Affiliation(s)
- Melissa Scholefield
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, UK.
| | - Stephanie J Church
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, UK.
| | - Sasha Philbert
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, UK.
| | - Jingshu Xu
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland, 1142, New Zealand.
| | - Stefano Patassini
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland, 1142, New Zealand.
| | - Garth J S Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, UK; School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland, 1142, New Zealand.
| |
Collapse
|
5
|
Amagai R, Otomo R, Yoshioka S, Nagano H, Hashimoto N, Sakakibara R, Tanaka T, Okado-Matsumoto A. C-terminal truncation is a prominent post-translational modification of human erythrocyte α-synuclein. J Biochem 2024; 175:649-658. [PMID: 38308089 DOI: 10.1093/jb/mvae012] [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: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
α-Synuclein (α-Syn) is a protein related to synucleinopathies with high expression in the central nervous system and erythrocytes which are a major source of peripheral α-Syn. Recent reports have suggested the presence of α-Syn within extracellular vesicles (EVs) derived from erythrocytes, potentially contributing to the pathogenesis of synucleinopathies. While Lewy bodies, intracellular inclusions containing aggregated α-Syn, are prominently observed within the brain, their occurrence in peripheral neurons implies the dissemination of synucleinopathy pathology throughout the body via the propagation of α-Syn. In this study, we found erythrocytes and circulating EVs obtained from plasma contained α-Syn, which was separated into four major forms using high-resolution clear native-PAGE and isoelectric focusing. Notably, erythrocyte α-Syn was classified into full-length and C-terminal truncated forms, with truncation observed between Y133 and Q134 as determined by LC-MS/MS analysis. Our finding revealed that C-terminally truncated α-Syn, which was previously reported to exist solely within the brain, was also present in erythrocytes and circulating EVs obtained from plasma.
Collapse
Affiliation(s)
- Ryosuke Amagai
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Riki Otomo
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Sakura Yoshioka
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Hidekazu Nagano
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Naoko Hashimoto
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ryuji Sakakibara
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Chiba 285-8741, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ayako Okado-Matsumoto
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
6
|
Zhang YN, Wu Q, Zhang NN, Chen HS. Ischemic Preconditioning Alleviates Cerebral Ischemia-Reperfusion Injury by Interfering With Glycocalyx. Transl Stroke Res 2023; 14:929-940. [PMID: 36168082 DOI: 10.1007/s12975-022-01081-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
Ischemic preconditioning (IPC) could protect the blood-brain barrier (BBB), but the underlying mechanism is not well understood. This preclinical study aimed to investigate whether glycocalyx could be involved in the neuroprotective effect of IPC on cerebral ischemia-reperfusion injury (IRI) and the possible mechanism in rat middle cerebral artery occlusion/reperfusion (MCAO/R) model. Neurological deficit scores, infarct volume, and brain edema were measured to assess the neuroprotection of IPC. Several serum biomarkers related to glycocalyx damage, such as hyaluronic acid (HA), heparan sulfate (HS), and syndecan-1 (SYND1), were evaluated, and their changes were normalized to the ratio of postoperative/preoperative concentration. Western blot and immunofluorescence were used to evaluate the content and cellular location of HA-related metabolic enzymes. This study found that (1) IPC improved brain infarction and edema, neurological impairment, and BBB disruption in IRI rats; (2) IPC significantly up-regulated HA ratio and down-regulated HS ratio, but did not affect SYND1 ratio compared with the IRI group. Moreover, the increased HA ratio was negatively related to brain edema and neurological deficit score. (3) IPC affected HA metabolism by up-regulating hyaluronate synthase-1 and matrix metalloproteinase-2, and down-regulating hyaluronidase-1 in brain tissue. Together, this is the first report that the neuroprotective effect of IPC on IRI may be mediated through interfering with glycocalyx in the MCAO/R model.
Collapse
Affiliation(s)
- Yi-Na Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
- Department of Neurology, General Hospital of Northern Theater Command of China Medical University, Shenyang, 110013, China
| | - Qiong Wu
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Nan-Nan Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China.
| |
Collapse
|
7
|
Saremi L, Esmaeili S, Ghaffari ME, Shahbazi S, Lotfipanah S, Kadkhodazadeh M. Evaluation of matrix metalloproteinase-1, -2, -3, -7, and -13 gene polymorphisms in patients with chronic periodontitis and healthy controls. Clin Oral Investig 2023; 27:7417-7423. [PMID: 37848583 DOI: 10.1007/s00784-023-05331-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES The current study aimed to investigate the association of matrix metalloproteinase- (MMP-) 1, -2, -3, -7, and -13 gene polymorphisms with chronic periodontitis (CP) in an Iranian population. MATERIALS AND METHODS In this case-control study, 87 subjects with CP and 89 periodontally healthy subjects were allocated to case and control groups, respectively. Subjects' venous blood samples (5 cc) were collected, and DNA extraction was performed. A spectrophotometer was utilized to assess the concentration of extracted DNAs. The desired gene polymorphisms were examined using restriction fragment length polymorphism polymerase chain reaction (RFLP-PCR) followed by electrophoresis. Statistical analyses were done using the Pearson Chi-Square test, odds ratio, and t-Test using SPSS Version 28. RESULTS The MMP-1 (-1607 1G/2G) rs1799750, MMP-3 (-1171 5A/6A) rs3025058, and MMP-7 (-181 A/G) rs11568818 gene polymorphisms significantly differed between case and control groups (PV = 0.019, 0.007, and 0.028, respectively). In contrast, the gene polymorphisms of MMP-2 (-1306 C/T) rs243865 and MMP-13 (-77 A/G) rs2252070 did not make a significant difference. Regarding allele frequencies, the presence of the 2G allele in the MMP-1 (-1607) rs1799750 genotype increased the CP susceptibility significantly, while subjects with the 6A allele in their MMP-3 (-1171) rs3025058 genotype showed significantly lower susceptibility to CP (PV = 0.008 and < 0.001, respectively). CONCLUSION In the studied population, gene polymorphisms in the DNA sequences of MMP-1 (-1607 1G/2G) rs1799750, MMP-3 (-1171 5A/6A) rs3025058, and MMP-7 (-181 A/G) rs11568818 may have impacts on CP incidence. CLINICAL RELEVANCE Clinicians should be cautious about the association between MMP-1, MMP-3, and MMP-7 gene polymorphisms and the incidence of chronic periodontitis during periodontal treatment planning.
Collapse
Affiliation(s)
- Leila Saremi
- Dental Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saharnaz Esmaeili
- Division of Oral and Maxillofacial Surgery, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
- Dentofacial Deformities Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ebrahim Ghaffari
- Department of Epidemiology and Biostatistics, Faculty of Health, Qom University of Medical Sciences, Qom, Iran
| | - Soheil Shahbazi
- Dentofacial Deformities Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Lotfipanah
- Department of Biology Education, Farhangian University, Tehran, Iran
| | - Mahdi Kadkhodazadeh
- Dental Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Rahman MM, Tumpa MAA, Rahaman MS, Islam F, Sutradhar PR, Ahmed M, Alghamdi BS, Hafeez A, Alexiou A, Perveen A, Ashraf GM. Emerging Promise of Therapeutic Approaches Targeting Mitochondria in Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:1081-1099. [PMID: 36927428 PMCID: PMC10286587 DOI: 10.2174/1570159x21666230316150559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 03/18/2023] Open
Abstract
Mitochondria are critical for homeostasis and metabolism in all cellular eukaryotes. Brain mitochondria are the primary source of fuel that supports many brain functions, including intracellular energy supply, cellular calcium regulation, regulation of limited cellular oxidative capacity, and control of cell death. Much evidence suggests that mitochondria play a central role in neurodegenerative disorders (NDDs) such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. Ongoing studies of NDDs have revealed that mitochondrial pathology is mainly found in inherited or irregular NDDs and is thought to be associated with the pathophysiological cycle of these disorders. Typical mitochondrial disturbances in NDDs include increased free radical production, decreased ATP synthesis, alterations in mitochondrial permeability, and mitochondrial DNA damage. The main objective of this review is to highlight the basic mitochondrial problems that occur in NDDs and discuss the use mitochondrial drugs, especially mitochondrial antioxidants, mitochondrial permeability transition blockade, and mitochondrial gene therapy, for the treatment and control of NDDs.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst. Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Popy Rani Sutradhar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Wien, Austria
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
9
|
Chloride Intracellular Channel Protein 2 Promotes Microglial Invasion: A Link to Microgliosis in the Parkinson's Disease Brain. Brain Sci 2022; 13:brainsci13010055. [PMID: 36672037 PMCID: PMC9857073 DOI: 10.3390/brainsci13010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Activated microglia potentially cause neurodegeneration in Parkinson's disease (PD). Matrix metalloproteinase (MMP)-9 plays a crucial role in the pathogenesis of PD, but the modulator of microglial release of MMP-9 remains obscure. Given the modulatory effect of chloride intracellular channel protein 2 (CLIC2) on MMPs, we aimed to determine the role of CLIC2 in regulating microglial MMP expression and activation. We found that CLIC2 is expressed in microglia and neurons in rat brain tissue and focused on the function of CLIC2 in primary cultured microglia. Exposure to recombinant CLIC2 protein enhanced microglial invasion activity, and its knockdown abolished this activity. Moreover, increased activation of MMP-9 was confirmed by the addition of the CLIC2 protein, and CLIC2 knockdown eliminated this activation. Additionally, increased expression of CLIC2 was observed in PD-modeled tissue. In conclusion, CLIC2 increases MMP-9 activity in the microglia, which are involved in PD pathogenesis.
Collapse
|
10
|
Bronisz E, Cudna A, Wierzbicka A, Kurkowska-Jastrzębska I. Serum Proteins Associated with Blood-Brain Barrier as Potential Biomarkers for Seizure Prediction. Int J Mol Sci 2022; 23:ijms232314712. [PMID: 36499038 PMCID: PMC9740683 DOI: 10.3390/ijms232314712] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
As 30% of epileptic patients remain drug-resistant, seizure prediction is vital. Induction of epileptic seizure is a complex process that can depend on factors such as intrinsic neuronal excitability, changes in extracellular ion concentration, glial cell activity, presence of inflammation and activation of the blood−brain barrier (BBB). In this study, we aimed to assess if levels of serum proteins associated with BBB can predict seizures. Serum levels of MMP-9, MMP-2, TIMP-1, TIMP-2, S100B, CCL-2, ICAM-1, P-selectin, and TSP-2 were examined in a group of 49 patients with epilepsy who were seizure-free for a minimum of seven days and measured by ELISA. The examination was repeated after 12 months. An extensive medical history was taken, and patients were subjected to a follow-up, including a detailed history of seizures. Serum levels of MMP-2, MMP-9, TIMP-1, CCL-2, and P-selectin differed between the two time points (p < 0.0001, p < 0.0001, p < 0.0001, p < 0.0001, p = 0.0035, respectively). General linear model analyses determined the predictors of seizures. Levels of MMP-2, MMP-9, and CCL-2 were found to influence seizure count in 1, 3, 6, and 12 months of observation. Serum levels of MMP-2, MMP-9, and CCL-2 may be considered potential biomarkers for seizure prediction and may indicate BBB activation.
Collapse
Affiliation(s)
- Elżbieta Bronisz
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
- Correspondence:
| | - Agnieszka Cudna
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Aleksandra Wierzbicka
- Sleep Disorders Center, Department of Clinical Neurophysiology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | | |
Collapse
|
11
|
Sánchez KE, Bhaskar K, Rosenberg GA. Apoptosis-associated speck-like protein containing a CARD-mediated release of matrix metalloproteinase 10 stimulates a change in microglia phenotype. Front Mol Neurosci 2022; 15:976108. [PMID: 36305000 PMCID: PMC9595131 DOI: 10.3389/fnmol.2022.976108] [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: 06/23/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation contributes to amyloid-β and tau pathology in Alzheimer's disease (AD). Microglia facilitate an altered immune response that includes microgliosis, upregulation of inflammasome proteins, and elevation of matrix-metalloproteinases (MMPs). Studies of cerebrospinal fluid (CSF) and blood in dementia patients show upregulation of two potential biomarkers of inflammation at the cellular level, MMP10 and apoptosis-associated speck-like protein containing a CARD (ASC). However, little is known about their relationship in the context of brain inflammation. Therefore, we stimulated microglia cultures with purified insoluble ASC speck aggregates and MMP10 to elucidate their role. We found that ASC specks altered microglia shape and stimulated the release of MMP3 and MMP10. Furthermore, MMP10 stimulated microglia released additional MMP10 along with the inflammatory cytokines, tumor-necrosis factor-α (TNFα), Interleukin 6 (IL-6), and CXCL1 CXC motif chemokine ligand 1 (CXCL1). A broad-spectrum MMP inhibitor, GM6001, prevented TNFα release. With these results, we conclude that MMP10 and ASC specks act on microglial cells to propagate inflammation.
Collapse
Affiliation(s)
- Kathryn E. Sánchez
- Center for Memory and Aging, University of New Mexico, Albuquerque, NM, United States
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, United States
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Gary A. Rosenberg
- Center for Memory and Aging, University of New Mexico, Albuquerque, NM, United States
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
12
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
13
|
Liu CZ, Guo DS, Ma JJ, Dong LR, Chang QQ, Yang HQ, Liang KK, Li XH, Yang DW, Fan YY, Gu Q, Chen SY, Li DS. Correlation of matrix metalloproteinase 3 and matrix metalloproteinase 9 levels with non-motor symptoms in patients with Parkinson’s disease. Front Aging Neurosci 2022; 14:889257. [PMID: 36072482 PMCID: PMC9444063 DOI: 10.3389/fnagi.2022.889257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Matrix metalloproteinases (MMPs) are essential for tissue formation, neuronal network remodeling, and blood–brain barrier integrity. MMPs have been widely studied in acute brain diseases. However, the relationship with Parkinson’s disease (PD) remains unclear. The purpose of this study was to evaluate the serum MMP3 and MMP9 levels of PD patients and analyze their correlation with non-motor symptoms. Methods In this cross-sectional study, we recruited 73 patients with idiopathic PD and 64 healthy volunteers. Serum MMP3 and MMP9 levels were measured by enzyme-linked immunosorbent assay (ELISA). Patients with PD were assessed for non-motor symptoms using the Non-motor Symptoms Scale (NMSS) and Parkinson’s disease sleep scale (PDSS) and Mini Mental State Examination (MMSE). Results Serum MMP3 levels were significantly decreased in PD patients, predominantly those with early-stage PD, compared with controls [12.56 (9.30, 17.44) vs. 15.37 (11.33, 24.41) ng/ml; P = 0.004], and the serum MMP9 levels of PD patients were significantly higher than those of healthy controls [522 (419, 729) vs. 329 (229, 473) ng/ml; P < 0.001]. MMP3 levels were positively correlated with the NMSS total score (r = 0.271, P = 0.020) and the single-item scores for item six, assessing the gastrointestinal tract (r = 0.333, P = 0.004), and there was an inverse correlation between serum MMP3 levels and PDSS score (r = –0.246, P = 0.036); meanwhile, MMP9 levels were positively correlated with the NMSS total score (r = 0.234, P = 0.047), and higher serum MMP9 levels were detected in the cognitive dysfunction subgroup than in the cognitively intact subgroup [658 (504, 877) vs. 502 (397, 608) ng/ml, P = 0.008]. Conclusion The serum MMP3 level of PD patients (especially early-stage patients) was significantly lower than that of the healthy control group, and the MMP9 level was significantly higher than that of the healthy control group. MMP3 and MMP9 levels correlate with sleep disturbance and cognitive function in PD patients, respectively.
Collapse
Affiliation(s)
- Chuan Ze Liu
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Da Shuai Guo
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jian Jun Ma
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
- *Correspondence: Jian Jun Ma,
| | - Lin Rui Dong
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qing Qing Chang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Hong Qi Yang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Ke Ke Liang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiao Huan Li
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Da Wei Yang
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Yong Yan Fan
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Qi Gu
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Si Yuan Chen
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Dong Sheng Li
- Department of Neurology, Henan University People’s Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Neurology, Zhengzhou University People’s Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Fakhri S, Sabouri S, Kiani A, Farzaei MH, Rashidi K, Mohammadi-Farani A, Mohammadi-Noori E, Abbaszadeh F. Intrathecal administration of naringenin improves motor dysfunction and neuropathic pain following compression spinal cord injury in rats: relevance to its antioxidant and anti-inflammatory activities. Korean J Pain 2022; 35:291-302. [PMID: 35768984 PMCID: PMC9251389 DOI: 10.3344/kjp.2022.35.3.291] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background Spinal cord injury (SCI) is one of the most debilitating disorders throughout the world, causing persistent sensory-motor dysfunction, with no effective treatment. Oxidative stress and inflammatory responses play key roles in the secondary phase of SCI. Naringenin (NAR) is a natural flavonoid with known anti-inflammatory and antioxidative properties. This study aims at evaluating the effects of intrathecal NAR administration on sensory-motor disability after SCI. Methods Animals underwent a severe compression injury using an aneurysm clip. About 30 minutes after surgery, NAR was injected intrathecally at the doses of 5, 10, and 15 mM in 20 µL volumes. For the assessment of neuropathic pain and locomotor function, acetone drop, hot plate, inclined plane, and Basso, Beattie, Bresnahan tests were carried out weekly till day 28 post-SCI. Effects of NAR on matrix metalloproteinase (MMP)-2 and MMP-9 activity was appraised by gelatin zymography. Also, histopathological analyses and serum levels of glutathione (GSH), catalase and nitrite were measured in different groups. Results NAR reduced neuropathic pain, improved locomotor function, and also attenuated SCI-induced weight loss weekly till day 28 post-SCI. Zymography analysis showed that NAR suppressed MMP-9 activity, whereas it increased that of MMP-2, indicating its anti-neuroinflammatory effects. Also, intrathecal NAR modified oxidative stress related markers GSH, catalase, and nitrite levels. Besides, the neuroprotective effect of NAR was corroborated through increased survival of sensory and motor neurons after SCI. Conclusions These results suggest intrathecal NAR as a promising candidate for medical therapeutics for SCI-induced sensory and motor dysfunction.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahryar Sabouri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Mohammadi-Farani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Mohammadi-Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.,Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
16
|
Acera A, Gómez-Esteban JC, Murueta-Goyena A, Galdos M, Azkargorta M, Elortza F, Ruzafa N, Ibarrondo O, Pereiro X, Vecino E. Potential Tear Biomarkers for the Diagnosis of Parkinson’s Disease—A Pilot Study. Proteomes 2022; 10:proteomes10010004. [PMID: 35076620 PMCID: PMC8788479 DOI: 10.3390/proteomes10010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease after Alzheimer’s disease. In this study, the tear proteome profile of patients with idiopathic PD (iPD, n = 24), carriers of the E46K-SNCA mutation (n = 3) and healthy control (CT, n = 27) subjects was analyzed to identify candidate biomarkers for the diagnosis of PD. An observational, prospective and case-control pilot study was carried out, analyzing the participants tear samples by nano-liquid chromatography–mass spectrometry (nLC–MS/MS) and assessing their neurological impairment. The proteomic data obtained are available at ProteomeXchange with identifier 10.6019/PXD028811. These analyses led to the identification of 560 tear proteins, some of which were deregulated in PD patients and that have been implicated in immune responses, inflammation, apoptosis, collagen degradation, protein synthesis, defense, lipid transport and altered lysosomal function. Of these proteins, six were related to neurodegenerative processes and showed a good capacity to classify patients and controls. These findings revealed that certain proteins were upregulated in the tears of PD patients, mainly proteins involved in lysosomal function. Thus, in this study, tear proteins were identified that are implicated in neurodegeneration and that may be related to an aggressive disease phenotype in PD patients.
Collapse
Affiliation(s)
- Arantxa Acera
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.A.); (N.R.); (X.P.)
- Department of Neuroscience, Biodonostia Health Research Institute, 20014 Donostia-San Sebastian, Spain
| | - Juan Carlos Gómez-Esteban
- Department of Neurology, Cruces University Hospital, 48903 Barakaldo, Spain;
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Ane Murueta-Goyena
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Marta Galdos
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
| | - Mikel Azkargorta
- Department of Proteomics, Center for Cooperative Research in Biosciences, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Felix Elortza
- Department of Proteomics, Center for Cooperative Research in Biosciences, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Noelia Ruzafa
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.A.); (N.R.); (X.P.)
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
| | | | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.A.); (N.R.); (X.P.)
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (A.A.); (N.R.); (X.P.)
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain;
- Correspondence: ; Tel.: +34-94-601-2820
| |
Collapse
|
17
|
Fu KA, Paul KC, Lu AT, Horvath S, Keener AM, Bordelon Y, Bronstein JM, Ritz B. DNA methylation-based surrogates of plasma proteins are associated with Parkinson's disease risk. J Neurol Sci 2021; 431:120046. [PMID: 34768133 DOI: 10.1016/j.jns.2021.120046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/24/2021] [Accepted: 11/01/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND The epigenome may reflect Parkinson's disease (PD) risk, which serves as a point of convergence of genetic and environmental risk factors. Here, we investigate whether blood DNA methylation (DNAm) markers are associated with PD risk. METHODS We selected 12 plasma proteins known as predictors of cardiovascular conditions and mortality to evaluate their effects on PD risk in a case-control study. In lieu of protein level measures, however, we assessed the influence of their DNAm surrogates. Primary analysis was restricted to 569 PD patients and 238 controls with DNAm data available. Using univariate logistic regression, we evaluated associations between the DNAm markers and PD. RESULTS Of the 12 DNAm surrogates, the most robustly associated were DNAm EFEMP-1 and DNAm CD56, which were associated with PD with and without controlling for blood cell composition. DNAm EFEMP-1 was associated with a decreased risk of PD (OR = 0.83 per SD, 95% CI = 0.70, 0.98) whereas DNAm CD56 was associated with an increased risk of PD (OR = 1.41, 95% CI = 1.11, 1.79). CONCLUSIONS Several DNAm markers, selected as part of a panel to track cardiovascular outcomes and mortality, were associated with PD risk. DNAm markers may inform of factors that are affected differentially in early PD patients compared with controls.
Collapse
Affiliation(s)
- Katherine A Fu
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| | - Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Ake T Lu
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Steve Horvath
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Biostatistics, UCLA Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Adrienne M Keener
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Neurology, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Yvette Bordelon
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Jeff M Bronstein
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Beate Ritz
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA.
| |
Collapse
|
18
|
Metformin and fluoxetine improve depressive-like behavior in a murine model of Parkinsońs disease through the modulation of neuroinflammation, neurogenesis and neuroplasticity. Int Immunopharmacol 2021; 102:108415. [PMID: 34890997 DOI: 10.1016/j.intimp.2021.108415] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/01/2023]
Abstract
Thereabout 30-40% of patients with Parkinson's Disease (PD) also have depression contributing to the loss of quality of life. Among the patients who treat depression, about 50% do not show significant improvement due to the limited efficacy of the treatment. So far, there are no effective disease-modifying treatments that can impede its progression. The current clinical approach is based on symptom management. Nonetheless, the reuse of drugs with excellent safety profiles represents an attractive alternative strategy for treating of different clinical aspects of PD. In this study, we evaluated the effects of metformin separately and associated with fluoxetine on depressive like-behavior and motor alterations in experimental Parkinson's disease. C57BL6 mice were induced with rotenone (2.5 mg/kg/day) for 20 days and treated with metformin (200 mg/kg/day) and fluoxetine (10 mg/kg/day) from the 5th day of induction. The animals were submitted to Sucrose Preference, Tail Suspension, and rotarod tests. Hippocampus, prefrontal cortex, and substantia nigra were dissected for molecular and morphological analysis. Metformin and fluoxetine prevented depressive-like behavior and improved motor impairment and increased TH nigral positive cells. Metformin and fluoxetine also reduced IBA-1 and GFAP positive cells in the hippocampus. Moreover, metformin reduced the phospho-NF-kB, IL-1β in the prefrontal cortex and iNOS levels in the hippocampus. Both metformin and fluoxetine increased neurogenesis by increasing KI67, but only the combined treatment increased neuronal survival by NeuN positive cells in the hippocampus. In addition, fluoxetine reduced cell death, decreasing caspase-3 and PARP-1 levels. Lastly, metformin potentiated the effect of fluoxetine on neuroplasticity by increasing BDNF positive cells. Metformin has antidepressant and antiparkinsonian potential due to anti-inflammatory neurogenic, and neuroplasticity-inducing effects when combined with fluoxetine.
Collapse
|
19
|
|
20
|
Choi H, Kim E, Choi JY, Park E, Lee HJ. Potent therapeutic targets for treatment of Alzheimer's disease: Amyloid degrading enzymes. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hang Choi
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Eungchan Kim
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Jae Yoon Choi
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| | - Eunsik Park
- Department of Life Sport Education Kongju National University Gongju Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education Kongju National University Gongju Republic of Korea
| |
Collapse
|
21
|
Kim T, Jeon J, Park JS, Park Y, Kim J, Noh H, Kim HS, Seo H. Matrix Metalloproteinase-8 Inhibitor Ameliorates Inflammatory Responses and Behavioral Deficits in LRRK2 G2019S Parkinson's Disease Model Mice. Biomol Ther (Seoul) 2021; 29:483-491. [PMID: 34045367 PMCID: PMC8411029 DOI: 10.4062/biomolther.2020.181] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 11/14/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder that involves the loss of dopaminergic neurons in the substantia nigra (SN). Matrix metalloproteinases-8 (MMP-8), neutrophil collagenase, is a functional player in the progressive pathology of various inflammatory disorders. In this study, we administered an MMP-8 inhibitor (MMP-8i) in Leucine-rich repeat kinase 2 (LRRK2) G2019S transgenic mice, to determine the effects of MMP-8i on PD pathology. We observed a significant increase of ionized calcium-binding adapter molecule 1 (Iba1)-positive activated microglia in the striatum of LRRK2 G2019S mice compared to normal control mice, indicating enhanced neuro-inflammatory responses. The increased number of Iba1-positive activated microglia in LRRK2 G2019S PD mice was down-regulated by systemic administration of MMP-8i. Interestingly, this LRRK2 G2019S PD mice showed significantly reduced size of cell body area of tyrosine hydroxylase (TH) positive neurons in SN region and MMP-8i significantly recovered cellular atrophy shown in PD model indicating distinct neuro-protective effects of MMP-8i. Furthermore, MMP-8i administration markedly improved behavioral abnormalities of motor balancing coordination in rota-rod test in LRRK2 G2019S mice. These data suggest that MMP-8i attenuates the pathological symptoms of PD through anti-inflammatory processes.
Collapse
Affiliation(s)
- Taewoo Kim
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jeha Jeon
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jin-Sun Park
- Department of Molecular Medicine and Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Yeongwon Park
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jooeui Kim
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Haneul Noh
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
22
|
Bortolanza M, do Nascimento GC, Raisman-Vozari R, Del-Bel E. Doxycycline and its derivative, COL-3, decrease dyskinesia induced by l-DOPA in hemiparkinsonian rats. Br J Pharmacol 2021; 178:2595-2616. [PMID: 33751546 DOI: 10.1111/bph.15439] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/03/2020] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE l-DOPA-induced dyskinesia is a debilitating effect of treating Parkinson's disease with this drug. New therapeutic approaches that prevent or attenuate this side effect are needed. EXPERIMENTAL APPROACH Wistar adult male rats submitted to 6-hydroxydopamine-induced unilateral medial forebrain bundle lesion were treated with l-DOPA (p.o. 20 mg·kg-1 or s.c. 10 mg·kg-1 ) once a day for 14 days. After this period, we tested if doxycycline (40 mg·kg-1 , i.p.) and COL-3 (50 and 100 nmol, i.c.v.) could reverse l-DOPA-induced dyskinesia. In an additional experiment, doxycycline was administered together with l-DOPA to verify if it would prevent l-DOPA-induced dyskinesia development. KEY RESULTS A single injection of doxycycline or COL-3 attenuated l-DOPA-induced dyskinesia. Co-treatment with doxycycline from the first day of l-DOPA suppressed the onset of dyskinesia. The improved motor response after l-DOPA was not affected by doxycycline or COL-3. Doxycycline treatment was associated with decreased immunoreactivity of FosB, COX-2, the astroglial protein GFAP and the microglial protein OX-42, which were elevated in the basal ganglia of rats exhibiting dyskinesia. Doxycycline decreased metalloproteinase-2/-9 activity, metalloproteinase-3 expression and ROS production. Metalloproteinase-2/-9 activity and production of ROS in the basal ganglia of dyskinetic rats showed a significant correlation with the intensity of dyskinesia. CONCLUSION AND IMPLICATIONS The present study demonstrates the anti-dyskinetic potential of doxycycline and its analogue compound COL-3 in hemiparkinsonian rats. Given the long-established and safe clinical use of doxycycline, this study suggests that these drugs might be tested to reduce or prevent l-DOPA-induced dyskinesia in Parkinson's patients.
Collapse
Affiliation(s)
- Mariza Bortolanza
- Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Elaine Del-Bel
- Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
23
|
Li R, Li Y, Mu M, Yang B, Chen X, Lee WYW, Ke Y, Yung WH, Tang BZ, Bian L. Multifunctional Nanoprobe for the Delivery of Therapeutic siRNA and Real-Time Molecular Imaging of Parkinson's Disease Biomarkers. ACS APPLIED MATERIALS & INTERFACES 2021; 13:11609-11620. [PMID: 33683858 DOI: 10.1021/acsami.0c22112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Parkinson's disease (PD) has been recently associated with the excessive expression of matrix metalloproteinase 3 (MMP3). One of the major challenges in treating PD is to effectively detect and inhibit the early MMP3 activities to relieve the neural stress and inflammation responses. Previously, numerous upconversion nanoparticle (UCNP)-based nanoprobes have been designed for the detection of biomarkers in neurodegenerative diseases. To further improve the performance of the conventional nanoprobes, we introduced novel reporting units and integrated the therapeutic reagents to fabricate a theragnostic platform for PD and other neurodegenerative diseases. Here, we designed a multifunctional UCNP/aggregation-induced emission luminogen (AIEgen)-based nanoprobe to effectively detect the time-lapse MMP3 activities in the inflammatory catecholaminergic SH-SY5Y cells and simultaneously deliver the MMP3-siRNA into the stressed catecholaminergic SH-SY5Y cells, inhibiting the MMP3-induced inflammatory neural responses. The unique features of our UCNP/AIEgen-based nanoprobe platform shed light on the development of a novel theragnostic probe for the early diagnosis and cure of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Yi Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Mingdao Mu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Xiaoyu Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Wayne Yuk Wai Lee
- Department of Orthopedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Wing Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
| | - Ben Zhong Tang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 999077 Hong Kong, P. R. China
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong, P. R. China
- The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen 518000, China
| |
Collapse
|
24
|
García-Bermúdez MY, Freude KK, Mouhammad ZA, van Wijngaarden P, Martin KK, Kolko M. Glial Cells in Glaucoma: Friends, Foes, and Potential Therapeutic Targets. Front Neurol 2021; 12:624983. [PMID: 33796062 PMCID: PMC8007906 DOI: 10.3389/fneur.2021.624983] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Glaucoma is the second leading cause of blindness worldwide, affecting ~80 million people by 2020 (1, 2). The condition is characterized by a progressive loss of retinal ganglion cells (RGCs) and their axons accompanied by visual field loss. The underlying pathophysiology of glaucoma remains elusive. Glaucoma is recognized as a multifactorial disease, and lowering intraocular pressure (IOP) is the only treatment that has been shown to slow the progression of the condition. However, a significant number of glaucoma patients continue to go blind despite intraocular pressure-lowering treatment (2). Thus, the need for alternative treatment strategies is indisputable. Accumulating evidence suggests that glial cells play a significant role in supporting RGC function and that glial dysfunction may contribute to optic nerve disease. Here, we review recent advances in understanding the role of glial cells in the pathophysiology of glaucoma. A particular focus is on the dynamic and essential interactions between glial cells and RGCs and potential therapeutic approaches to glaucoma by targeting glial cells.
Collapse
Affiliation(s)
| | - Kristine K Freude
- Department for Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Zaynab A Mouhammad
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Peter van Wijngaarden
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Keith K Martin
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| |
Collapse
|
25
|
Morató X, Garcia-Esparcia P, Argerich J, Llorens F, Zerr I, Paslawski W, Borràs E, Sabidó E, Petäjä-Repo UE, Fernández-Dueñas V, Ferrer I, Svenningsson P, Ciruela F. Ecto-GPR37: a potential biomarker for Parkinson's disease. Transl Neurodegener 2021; 10:8. [PMID: 33637132 PMCID: PMC7908677 DOI: 10.1186/s40035-021-00232-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/08/2021] [Indexed: 11/15/2022] Open
Abstract
Objective α-Synuclein has been studied as a potential biomarker for Parkinson’s disease (PD) with no concluding results. Accordingly, there is an urgent need to find out reliable specific biomarkers for PD. GPR37 is an orphan G protein-coupled receptor that toxically accumulates in autosomal recessive juvenile parkinsonism. Here, we investigated whether GPR37 is upregulated in sporadic PD, and thus a suitable potential biomarker for PD. Methods GPR37 protein density and mRNA expression in postmortem substantia nigra (SN) from PD patients were analysed by immunoblot and RT-qPCR, respectively. The presence of peptides from the N-terminus-cleaved domain of GPR37 (i.e. ecto-GPR37) in human cerebrospinal fluid (CSF) was determined by liquid chromatography-mass spectrometric analysis. An engineered in-house nanoluciferase-based immunoassay was used to quantify ecto-GPR37 in CSF samples from neurological control (NC) subjects, PD patients and Alzheimer’s disease (AD) patients. Results GPR37 protein density and mRNA expression were significantly augmented in sporadic PD. Increased amounts of ecto-GPR37 peptides in the CSF samples from PD patients were identified by mass spectrometry and quantified by the in-house ELISA method. However, the CSF total α-synuclein level in PD patients did not differ from that in NC subjects. Similarly, the cortical GPR37 mRNA expression and CSF ecto-GPR37 levels in AD patients were also unaltered. Conclusion GPR37 expression is increased in SN of sporadic PD patients. The ecto-GPR37 peptides are significantly increased in the CSF of PD patients, but not in AD patients. These results open perspectives and encourage further clinical studies to confirm the validity and utility of ecto-GPR37 as a potential PD biomarker. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-021-00232-7.
Collapse
Affiliation(s)
- Xavier Morató
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain.,Section of Neurology, Department of Clinical Neuroscience, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Paula Garcia-Esparcia
- Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain.,Neuropathology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Madrid, Spain
| | - Josep Argerich
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Franc Llorens
- Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain.,Neuropathology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Madrid, Spain.,Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Göttingen, Germany.,German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Wojciech Paslawski
- Section of Neurology, Department of Clinical Neuroscience, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Eva Borràs
- Proteomics Unit, Center for Genomic Regulation, Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Center for Genomic Regulation, Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ulla E Petäjä-Repo
- Research Unit of Biomedicine, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Víctor Fernández-Dueñas
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Isidro Ferrer
- Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain.,Neuropathology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Madrid, Spain
| | - Per Svenningsson
- Section of Neurology, Department of Clinical Neuroscience, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain. .,Neuroscience Program, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
26
|
Xue Y, Liang H, Yang R, Deng K, Tang M, Zhang M. The role of pro- and mature neurotrophins in the depression. Behav Brain Res 2021; 404:113162. [PMID: 33549684 DOI: 10.1016/j.bbr.2021.113162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
Neurotrophic factors, which can provide nutritional support to neurons and neuronal cells, also played an important role in their proliferation and survival. As signaling molecules, it also mediated the learning, memory and other activities in the brain. The latest study shows that neurotrophic factors have diametrically opposing effects of the pro- and mature form through distinct receptors. In this review, we summarize the different forms of neurotrophic factors, related receptors, and the corresponding biological effects. More importantly, we expounded the physiology and pathology mechanisms of brain-derived neurotrophic factor(BDNF)in depression. It is hopefully to provide new idea on the relationship of neurotrophic factors and depression.
Collapse
Affiliation(s)
- Ying Xue
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Hongyan Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Rui Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Kunhong Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
27
|
Behl T, Kaur G, Sehgal A, Bhardwaj S, Singh S, Buhas C, Judea-Pusta C, Uivarosan D, Munteanu MA, Bungau S. Multifaceted Role of Matrix Metalloproteinases in Neurodegenerative Diseases: Pathophysiological and Therapeutic Perspectives. Int J Mol Sci 2021; 22:ijms22031413. [PMID: 33573368 PMCID: PMC7866808 DOI: 10.3390/ijms22031413] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegeneration is the pathological condition, in which the nervous system or neuron loses its structure, function, or both, leading to progressive degeneration or the death of neurons, and well-defined associations of tissue system, resulting in clinical manifestations. Neuroinflammation has been shown to precede neurodegeneration in several neurodegenerative diseases (NDs). No drug is yet known to delay or treat neurodegeneration. Although the etiology and potential causes of NDs remain widely indefinable, matrix metalloproteinases (MMPs) evidently have a crucial role in the progression of NDs. MMPs, a protein family of zinc (Zn2+)-containing endopeptidases, are pivotal agents that are involved in various biological and pathological processes in the central nervous system (CNS). The current review delineates the several emerging evidence demonstrating the effects of MMPs in the progression of NDs, wherein they regulate several processes, such as (neuro)inflammation, microglial activation, amyloid peptide degradation, blood brain barrier (BBB) disruption, dopaminergic apoptosis, and α-synuclein modulation, leading to neurotoxicity and neuron death. Published papers to date were searched via PubMed, MEDLINE, etc., while using selective keywords highlighted in our manuscript. We also aim to shed a light on pathophysiological effect of MMPs in the CNS and focus our attention on its detrimental and beneficial effects in NDs, with a special focus on Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), multiple sclerosis (MS), and Huntington's disease (HD), and discussed various therapeutic strategies targeting MMPs, which could serve as potential modulators in NDs. Over time, several agents have been developed in order to overcome challenges and open up the possibilities for making selective modulators of MMPs to decipher the multifaceted functions of MMPs in NDs. There is still a greater need to explore them in clinics.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| | - Gagandeep Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Shaveta Bhardwaj
- Department of Pharmacology, GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana 141104, Punjab, India;
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Camelia Buhas
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (C.J.-P.)
| | - Claudia Judea-Pusta
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (C.J.-P.)
| | - Diana Uivarosan
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| |
Collapse
|
28
|
Perry EA, Bennett CF, Luo C, Balsa E, Jedrychowski M, O'Malley KE, Latorre-Muro P, Ladley RP, Reda K, Wright PM, Gygi SP, Myers AG, Puigserver P. Tetracyclines promote survival and fitness in mitochondrial disease models. Nat Metab 2021; 3:33-42. [PMID: 33462515 PMCID: PMC7856165 DOI: 10.1038/s42255-020-00334-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial diseases (MDs) are a heterogeneous group of disorders resulting from mutations in nuclear or mitochondrial DNA genes encoding mitochondrial proteins1,2. MDs cause pathologies with severe tissue damage and ultimately death3,4. There are no cures for MDs and current treatments are only palliative5-7. Here we show that tetracyclines improve fitness of cultured MD cells and ameliorate disease in a mouse model of Leigh syndrome. To identify small molecules that prevent cellular damage and death under nutrient stress conditions, we conduct a chemical high-throughput screen with cells carrying human MD mutations and discover a series of antibiotics that maintain survival of various MD cells. We subsequently show that a sub-library of tetracycline analogues, including doxycycline, rescues cell death and inflammatory signatures in mutant cells through partial and selective inhibition of mitochondrial translation, resulting in an ATF4-independent mitohormetic response. Doxycycline treatment strongly promotes fitness and survival of Ndufs4-/- mice, a preclinical Leigh syndrome mouse model8. A proteomic analysis of brain tissue reveals that doxycycline treatment largely prevents neuronal death and the accumulation of neuroimmune and inflammatory proteins in Ndufs4-/- mice, indicating a potential causal role for these proteins in the brain pathology. Our findings suggest that tetracyclines deserve further evaluation as potential drugs for the treatment of MDs.
Collapse
Affiliation(s)
- Elizabeth A Perry
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Biological Sciences in Dental Medicine Program, Harvard School of Dental Medicine, Boston, MA, USA
| | - Christopher F Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Chi Luo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Mark Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Katherine E O'Malley
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Richard Porter Ladley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Kamar Reda
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Peter M Wright
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Andrew G Myers
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Wang M, Xie Y, Qin D. Proteolytic cleavage of proBDNF to mBDNF in neuropsychiatric and neurodegenerative diseases. Brain Res Bull 2021; 166:172-184. [PMID: 33202257 DOI: 10.1016/j.brainresbull.2020.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is involved in pathophysiological mechanisms in neuropsychiatric diseases, including depression, anxiety, and schizophrenia (SZ), as well as neurodegenerative diseases like Parkinson's disease (PD) and Alzheimer's disease (AD). An imbalance or insufficient pro-brain-derived neurotrophic factor (proBDNF) transformation into mature BDNF (mBDNF) is potentially critical to the disease pathogenesis by impairing neuronal plasticity as suggested by results from many studies. Thus, promoting proBDNF transformation into mBDNF is therefore hypothesized as beneficial for the treatment of neuropsychiatric and neurodegenerative diseases. ProBDNF is proteolytically cleaved into the mBDNF by intracellular furin/proprotein convertases and extracellular proteases (plasmin/matrix metallopeptidases). This article reviews the mechanisms of the conversion of proBDNF to mBDNF and the research status of intracellular/extracellular proteolytic proteases for neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Mingyue Wang
- School of Traditional Chinese Pharmacy, Yunnan University of Chinese Medicine, Yunnan 650500, China
| | - Yuhuan Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Yunnan 650500, China.
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Yunnan 650500, China.
| |
Collapse
|
30
|
Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int J Mol Sci 2020; 21:E9739. [PMID: 33419373 PMCID: PMC7767220 DOI: 10.3390/ijms21249739] [Citation(s) in RCA: 866] [Impact Index Per Article: 173.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent extracellular matrix (ECM) remodeling endopeptidases that have the capacity to degrade almost every component of the ECM. The degradation of the ECM is of great importance, since it is related to embryonic development and angiogenesis. It is also involved in cell repair and the remodeling of tissues. When the expression of MMPs is altered, it can generate the abnormal degradation of the ECM. This is the initial cause of the development of chronic degenerative diseases and vascular complications generated by diabetes. In addition, this process has an association with neurodegeneration and cancer progression. Within the ECM, the tissue inhibitors of MMPs (TIMPs) inhibit the proteolytic activity of MMPs. TIMPs are important regulators of ECM turnover, tissue remodeling, and cellular behavior. Therefore, TIMPs (similar to MMPs) modulate angiogenesis, cell proliferation, and apoptosis. An interruption in the balance between MMPs and TIMPs has been implicated in the pathophysiology and progression of several diseases. This review focuses on the participation of both MMPs (e.g., MMP-2 and MMP-9) and TIMPs (e.g., TIMP-1 and TIMP-3) in physiological processes and on how their abnormal regulation is associated with human diseases. The inclusion of current strategies and mechanisms of MMP inhibition in the development of new therapies targeting MMPs was also considered.
Collapse
Affiliation(s)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| | | | | | | | | | | | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| |
Collapse
|
31
|
Sánchez K, Maguire-Zeiss K. MMP13 Expression Is Increased Following Mutant α-Synuclein Exposure and Promotes Inflammatory Responses in Microglia. Front Neurosci 2020; 14:585544. [PMID: 33343280 PMCID: PMC7738560 DOI: 10.3389/fnins.2020.585544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
α-Synuclein is a 140-amino acid protein that readily misfolds and is associated with the Lewy body pathology found in sporadic and genetic forms of Parkinson's disease. We and others have shown that wild-type α-synuclein is a damage-associated molecular pattern that directly elicits a proinflammatory response in microglia through toll-like receptor activation. Here we investigated the direct effect of oligomeric mutant α-synuclein (A53T) on microglia morphology and activation. We found that misfolded A53T increased quantitative measures of amoeboid cell morphology, NFκB nuclear translocation and the expression of prototypical proinflammatory molecules. We also demonstrated that A53T increased expression of MMP13, a matrix metalloproteinase that remodels the extracellular matrix. To better understand the role of MMP13 in synucleinopathies, we further characterized the role of MMP13 in microglial signaling. We showed exposure of microglia to MMP13 induced a change in morphology and promoted the release of TNFα and MMP9. Notably, IL1β was not released indicating that the pathway involved in MMP13 activation of microglia may be different than the A53T pathway. Lastly, MMP13 increased the expression of CD68 suggesting that the lysosomal pathway might be altered by this MMP. Taken together this study shows that mutant α-synuclein directly induces a proinflammatory phenotype in microglia, which includes the expression of MMP13. In turn, MMP13 directly alters microglia supporting the need for multi-target therapies to treat Parkinson's disease patients.
Collapse
Affiliation(s)
- Kathryn Sánchez
- Department of Biology, Georgetown University, Washington, DC, United States
| | - Kathleen Maguire-Zeiss
- Department of Biology, Georgetown University, Washington, DC, United States.,Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
32
|
Morello G, Salomone S, D’Agata V, Conforti FL, Cavallaro S. From Multi-Omics Approaches to Precision Medicine in Amyotrophic Lateral Sclerosis. Front Neurosci 2020; 14:577755. [PMID: 33192262 PMCID: PMC7661549 DOI: 10.3389/fnins.2020.577755] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and fatal neurodegenerative disorder, caused by the degeneration of upper and lower motor neurons for which there is no truly effective cure. The lack of successful treatments can be well explained by the complex and heterogeneous nature of ALS, with patients displaying widely distinct clinical features and progression patterns, and distinct molecular mechanisms underlying the phenotypic heterogeneity. Thus, stratifying ALS patients into consistent and clinically relevant subgroups can be of great value for the development of new precision diagnostics and targeted therapeutics for ALS patients. In the last years, the use and integration of high-throughput "omics" approaches have dramatically changed our thinking about ALS, improving our understanding of the complex molecular architecture of ALS, distinguishing distinct patient subtypes and providing a rational foundation for the discovery of biomarkers and new individualized treatments. In this review, we discuss the most significant contributions of omics technologies in unraveling the biological heterogeneity of ALS, highlighting how these approaches are revealing diagnostic, prognostic and therapeutic targets for future personalized interventions.
Collapse
Affiliation(s)
- Giovanna Morello
- Institute for Research and Biomedical Innovation (IRIB), Italian National Research Council (CNR), Catania, Italy
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Velia D’Agata
- Human Anatomy and Histology, University of Catania, Catania, Italy
| | | | - Sebastiano Cavallaro
- Institute for Research and Biomedical Innovation (IRIB), Italian National Research Council (CNR), Catania, Italy
| |
Collapse
|
33
|
Dyavar SR, Potts LF, Beck G, Dyavar Shetty BL, Lawson B, Podany AT, Fletcher CV, Amara RR, Papa SM. Transcriptomic approach predicts a major role for transforming growth factor beta type 1 pathway in L-Dopa-induced dyskinesia in parkinsonian rats. GENES BRAIN AND BEHAVIOR 2020; 19:e12690. [PMID: 32741046 DOI: 10.1111/gbb.12690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 01/21/2023]
Abstract
Dyskinesia induced by long-term L-Dopa (LID) therapy in Parkinson disease is associated with altered striatal function whose molecular bases remain unclear. Here, a transcriptomic approach was applied for comprehensive analysis of distinctively regulated genes in striatal tissue, their specific pathways, and functional- and disease-associated networks in a rodent model of LID. This approach has identified transforming growth factor beta type 1 (TGFβ1) as a highly upregulated gene in dyskinetic animals. TGFβ1 pathway is a top aberrantly regulated pathway in the striatum following LID development based on differentially expressed genes (> 1.5 fold change and P < 0.05). The induction of TGFβ1 pathway specific genes, TGFβ1, INHBA, AMHR2 and PMEPA1 was also associated with regulation of NPTX2, PDP1, SCG2, SYNPR, TAC1, TH, TNNT1 genes. Transcriptional network and upstream regulator analyses have identified AKT-centered functional and ERK-centered disease networks revealing the association of TGFβ1, IL-1β and TNFα with LID development. Therefore, results support that TGFβ1 pathway is a major contributor to the pathogenic mechanisms of LID.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Lisa F Potts
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Goichi Beck
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | | | - Benton Lawson
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Anthony T Podany
- Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Courtney V Fletcher
- Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rama Rao Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
34
|
Omori W, Hattori K, Kajitani N, Okada-Tsuchioka M, Boku S, Kunugi H, Okamoto Y, Takebayashi M. Increased matrix metalloproteinases in cerebrospinal fluids of patients with major depressive disorder and schizophrenia. Int J Neuropsychopharmacol 2020; 23:pyaa049. [PMID: 32671384 PMCID: PMC7745248 DOI: 10.1093/ijnp/pyaa049] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 07/10/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chronic inflammation of the brain has a pivotal role in the pathophysiology of major depressive disorder (MDD) and schizophrenia (SCZ). Matrix metalloproteinases (MMPs) are extracellular proteases involved in pro-inflammatory processes and interact with IL-6, which is increased in the cerebrospinal fluid (CSF) of patients with MDD and SCZ. However, MMPs in the CSF in patients with MDD and SCZ remains unclear. Therefore, we compared MMPs in the CSF of patients with MDD and SCZ to those of healthy controls (HC). METHODS Japanese patients were diagnosed with DSM-IV-TR and clinical symptoms were assessed with the Hamilton Rating Scale for Depression for MDD and the Positive and Negative Syndrome Scale for SCZ. CSF was obtained from MDD (n=90), SCZ (n=86) and from age- and sex-matched HC (n=106). The levels of MMPs in CSF were measured with multiplex bead-based immunoassay. RESULTS The levels of MMP-2 in CSF were higher in both MDD and SCZ than HC and were positively correlated with clinical symptomatic scores in MDD, but not in SCZ. Regardless of diagnosis, the levels of MMP-2, -7 and -10 were positively correlated with each other, and the levels of MMP-7 and -10 were higher in MDD, but not in SCZ, compared to HC. CONCLUSION Increased CSF levels of MMP-2 in MDD and SCZ may be associated with brain inflammation. State-dependent alteration of MMP-2 and activation of cascades involving MMP-2, -7, and -10 appeared to have a role in the pathophysiology of MDD.
Collapse
Affiliation(s)
- Wataru Omori
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima, Japan
- Department of Psychiatry, NHO Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima, Japan
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Naoto Kajitani
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima, Japan
- Department of Neuropsychiatry, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| | - Mami Okada-Tsuchioka
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima, Japan
| | - Shuken Boku
- Department of Neuropsychiatry, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo, Japan
| | - Yasumasa Okamoto
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Minoru Takebayashi
- Division of Psychiatry and Neuroscience, Institute for Clinical Research, National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center, Kure, Hiroshima, Japan
- Department of Neuropsychiatry, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
35
|
Shin JY, Lee PH. Mesenchymal stem cells modulate misfolded α-synuclein in parkinsonian disorders: A multitarget disease-modifying strategy. Stem Cell Res 2020; 47:101908. [PMID: 32683319 DOI: 10.1016/j.scr.2020.101908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/22/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-Syn) aggregates, the major toxic component of Lewy bodies, are proteinaceous fibrillar cytoplasmic inclusions observed in α-synucleinopathies, such as Parkinson's disease (PD), multiple system atrophy, and dementia with Lewy bodies. Overexpression of α-syn induce neuronal loss and α-syn aggregation in PD animals. Recent studies show that α-syn is released by exocytosis and can be transmitted between brain areas through cell-to-cell propagation. Moreover, aggregates of extracellular α-syn can induce neuroinflammation-mediated neurotoxic signaling through microglial activation and release of pro-inflammatory factors. Thus, modulation of α-syn might be a potential therapeutic strategy for modifying disease progression of α-synucleinopathies. Our previous studies have revealed that MSCs have potent neuroprotective effects in PD animal through modulation of neuroinflammation, inhibition of cell death, and promotion of neurogenesis. Here, we provide further evidence that MSCs have the potential to modulate α-syn-related microenvironments via enhancement of autophagy, proteolysis of α-syn aggregates, inhibition of cell-to-cell transmission of α-syn, stabilization of axonal transport, and phagocytic clearance of α-syn by microglial M2 polarization. With advantages in clinical applications, these data suggests that the use of MSCs as pharmacological modulators of α-syn propagation would be an effective therapeutic approach in PD.
Collapse
Affiliation(s)
- Jin Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea.
| |
Collapse
|
36
|
Costa T, Fernandez-Villalba E, Izura V, Lucas-Ochoa AM, Menezes-Filho NJ, Santana RC, de Oliveira MD, Araújo FM, Estrada C, Silva V, Costa SL, Herrero MT. Combined 1-Deoxynojirimycin and Ibuprofen Treatment Decreases Microglial Activation, Phagocytosis and Dopaminergic Degeneration in MPTP-Treated Mice. J Neuroimmune Pharmacol 2020; 16:390-402. [PMID: 32564332 DOI: 10.1007/s11481-020-09925-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/14/2020] [Indexed: 12/28/2022]
Abstract
Inflammation is a predominant aspect of neurodegenerative diseases and experimental studies performed in animal models of Parkinson's disease (PD) suggesting that a sustained neuroinflammation exacerbates the nigrostriatal degeneration pathway. The central role of microglia in neuroinflammation has been studied as a target for potential neuroprotective drugs for PD, for example nonsteroidal anti-inflammatory drugs (NSAIDs) and matrix metalloproteinases (MMP) inhibitors that regulates microglial activation and migration. The aim of this study was to investigate the neuroprotective response of the iminosugar 1-deoxynojirimycin (1-DNJ) and compare its effect with a combined treatment with ibuprofen. MPTP-treated mice were orally dosed with ibuprofen and/or 1-DNJ 1. Open-field test was used to evaluate behavioral changes. Immunohistochemistry for dopaminergic neurons marker (TH+) and microglia markers (Iba-1+; CD68+) were used to investigate neuronal integrity and microglial activation in the substantia nigra pars compacta (SNpc). The pro-inflammatory cytokines TNF-α and IL-6 were analysed by qPCR. Treatments with either 1-DNJ or Ibuprofen alone did not reduce the damage induced by MPTP intoxication. However, combined treatment with 1-DNJ and ibuprofen prevents loss of mesencephalic dopaminergic neurons, decreases the number of CD68+/ Iba-1+ cells, the microglia/neurons interactions, and the pro-inflammatory cytokines, and improves behavioral changes when compared with MPTP-treated animals. In conclusion, these data demonstrate that the combined treatment with a MMPs inhibitor (1-DNJ) plus an anti-inflammatory drug (ibuprofen) has neuroprotective effects open for future therapeutic interventions. Graphical Abstract MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a protoxicant that, after crossing the Blood Brain Barrier, is metabolized by astrocytic MAO-B to MPDP+, a pyridinium intermediate, which undergoes further two-electron oxidation to yield the toxic metabolite MPP+ (methyl-phenyltetrahydropyridinium) that is then selectively transported into nigral neurons via the mesencephalic dopamine transporter. In this study, we demonstrated that MPTP induced death of dopaminergic neurons, microgliosis, increase of gliapses, motor impairment and neuroinflammation in mice, which were inhibited by combined 1-deoxynojirimycin and ibuprofen treatment.
Collapse
Affiliation(s)
- Tcs Costa
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain.,Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - E Fernandez-Villalba
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain
| | - V Izura
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain
| | - A M Lucas-Ochoa
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain
| | - N J Menezes-Filho
- Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - R C Santana
- Department of Bioregulation, Laboratory of Neuroscience, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - M D de Oliveira
- Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.,Faculty of Ceilandia, University of Brasilia - UnB, Brasilia, Federal District, Brazil
| | - F M Araújo
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain.,Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - C Estrada
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain
| | - Vda Silva
- Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil
| | - S L Costa
- Department of Biochemistry and Biophysics, Laboratory of Neurochemistry and Cell Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-100, Brazil.
| | - M T Herrero
- Clinical & Experimental Neuroscience (NiCE). Institute for Bio-Health Research of Murcia (IMIB), Institute for Aging Research (IUIE). School of Medicine, University of Murcia, Murcia, Spain.
| |
Collapse
|
37
|
Boguszewska-Czubara A, Budzynska B, Skalicka-Wozniak K, Kurzepa J. Perspectives and New Aspects of Metalloproteinases' Inhibitors in the Therapy of CNS Disorders: From Chemistry to Medicine. Curr Med Chem 2019; 26:3208-3224. [PMID: 29756562 DOI: 10.2174/0929867325666180514111500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/31/2017] [Accepted: 04/05/2018] [Indexed: 11/22/2022]
Abstract
Matrix metalloproteinases (MMPs) play a key role in remodeling of the extracellular matrix (ECM) and, at the same time, influence cell differentiation, migration, proliferation, and survival. Their importance in a variety of human diseases including cancer, rheumatoid arthritis, pulmonary emphysema and fibrotic disorders has been known for many years but special attention should be paid on the role of MMPs in the central nervous system (CNS) disorders. Till now, there are not many well documented physiological MMP target proteins in the brain but only some pathological ones. Numerous neurodegenerative diseases are a consequence of or result in disturbed remodeling of brain ECM, therefore proper action of MMPs as well as control of their activity may play crucial roles in the development of these diseases. In the present review, we discuss the role of metalloproteinase inhibitors, from the wellknown natural endogenous tissue inhibitors of metalloproteinases (TIMPs) to the exogenous synthetic ones like (4-phenoxyphenylsulfonyl)methylthiirane (SB-3CT), tetracyclines, batimastat (BB-94) and FN-439. As the MMP-TIMP system has been well described in physiological development as well as in pathological conditions mainly in neoplastic diseases, the knowledge about the enzymatic system in mammalian brain tissue still remains poorly understood in this context. Therefore, we focus on MMPs inhibition in the context of the physiological function of the adult brain as well as pathological conditions including neurodegenerative diseases, brain injuries, and others.
Collapse
Affiliation(s)
| | - Barbara Budzynska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin, Poland
| | - Krystyna Skalicka-Wozniak
- Department of Pharmacognosy with Medicinal Plants Unit, Medical University of Lublin, Lublin, Poland
| | - Jacek Kurzepa
- Department of Medical Chemistry, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
38
|
Spindola A, Targa ADS, Rodrigues LS, Winnischofer SMB, Lima MMS, Sogayar MC, Trombetta-Lima M. Increased Mmp/Reck Expression Ratio Is Associated with Increased Recognition Memory Performance in a Parkinson's Disease Animal Model. Mol Neurobiol 2019; 57:837-847. [PMID: 31493243 DOI: 10.1007/s12035-019-01740-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/19/2019] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide. Among its non-motor symptoms, sleep disorders are extremely common, being linked to cognitive and memory disruption. The microenvironment, particularly the extracellular matrix (ECM), is deeply involved in memory consolidation as well as in neuropathological processes, such as inflammation, damage to the blood-brain barrier and neuronal death. To better understand ECM dynamics in PD memory disturbances, we investigated the orchestrated expression of Mmps (Mmp-3, Mmp-7, and Mmp-9) and their modulators (Reck and Timp-3) in a rotenone-induced PD model. Also, we introduced an additional intervention in the memory process through rapid eye movement sleep deprivation (REMSD). We observed a REMSD-induced trend in reversing the memory impairment caused by rotenone administration. Associated to this phenotype, we observed a significant increase in Mmp-7/Reck and Mmp-9/Reck mRNA expression ratio in the substantia nigra and Mmp-9/Reck ratio in the hypothalamus. Moreover, the positive correlation of Mmp/Reck expression ratios between the substantia nigra and the striatum, observed upon rotenone infusion, was reversed by REMSD. Taken together, our results suggest a potential orchestrated association between an increase in Mmp-7 and Mmp-9/Reck expression ratios in the substantia nigra and a possible positive effect on cognitive performance in subjects affected by PD.
Collapse
Affiliation(s)
- Adauto Spindola
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP, 05360-130, Brazil
| | - Adriano D S Targa
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil.,Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil
| | - Lais Soares Rodrigues
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil.,Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil
| | - Sheila Maria Brochado Winnischofer
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil.,Departamento de Biologia Celular e Molecular, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil
| | - Marcelo M S Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil.,Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, 81531-990, Brazil
| | - Mari Cleide Sogayar
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP, 05360-130, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Marina Trombetta-Lima
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, SP, 05360-130, Brazil.
| |
Collapse
|
39
|
Rivera S, García-González L, Khrestchatisky M, Baranger K. Metalloproteinases and their tissue inhibitors in Alzheimer's disease and other neurodegenerative disorders. Cell Mol Life Sci 2019; 76:3167-3191. [PMID: 31197405 PMCID: PMC11105182 DOI: 10.1007/s00018-019-03178-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
As life expectancy increases worldwide, age-related neurodegenerative diseases will increase in parallel. The lack of effective treatment strategies may soon lead to an unprecedented health, social and economic crisis. Any attempt to halt the progression of these diseases requires a thorough knowledge of the pathophysiological mechanisms involved to facilitate the identification of new targets and the application of innovative therapeutic strategies. The metzincin superfamily of metalloproteinases includes matrix metalloproteinases (MMP), a disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS). These multigenic and multifunctional proteinase families regulate the functions of an increasing number of signalling and scaffolding molecules involved in neuroinflammation, blood-brain barrier disruption, protein misfolding, synaptic dysfunction or neuronal death. Metalloproteinases and their physiological inhibitors, the tissue inhibitors of metalloproteinases (TIMPs), are therefore, at the crossroads of molecular and cellular mechanisms that support neurodegenerative processes, and emerge as potential new therapeutic targets. We provide an overview of current knowledge on the role and regulation of metalloproteinases and TIMPs in four major neurodegenerative diseases: Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington's disease.
Collapse
Affiliation(s)
- Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| | | | | | - Kévin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| |
Collapse
|
40
|
Booth HDE, Wessely F, Connor-Robson N, Rinaldi F, Vowles J, Browne C, Evetts SG, Hu MT, Cowley SA, Webber C, Wade-Martins R. RNA sequencing reveals MMP2 and TGFB1 downregulation in LRRK2 G2019S Parkinson's iPSC-derived astrocytes. Neurobiol Dis 2019; 129:56-66. [PMID: 31085228 DOI: 10.1016/j.nbd.2019.05.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/09/2019] [Accepted: 05/10/2019] [Indexed: 12/26/2022] Open
Abstract
Non-neuronal cell types such as astrocytes can contribute to Parkinson's disease (PD) pathology. The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is one of the most common known causes of familial PD. To characterize its effect on astrocytes, we developed a protocol to produce midbrain-patterned astrocytes from human induced pluripotent stem cells (iPSCs) derived from PD LRRK2 G2019S patients and healthy controls. RNA sequencing analysis revealed the downregulation of genes involved in the extracellular matrix in PD cases. In particular, transforming growth factor beta 1 (TGFB1), which has been shown to inhibit microglial inflammatory response in a rat model of PD, and matrix metallopeptidase 2 (MMP2), which has been shown to degrade α-synuclein aggregates, were found to be down-regulated in LRRK2 G2019S astrocytes. Our findings suggest that midbrain astrocytes carrying the LRRK2 G2019S mutation may have reduced neuroprotective capacity and may contribute to the development of PD pathology.
Collapse
Affiliation(s)
| | - Frank Wessely
- UK Dementia Research Institute, Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK
| | | | - Federica Rinaldi
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Cathy Browne
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Samuel G Evetts
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michele T Hu
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sally A Cowley
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Caleb Webber
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; UK Dementia Research Institute, Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK.
| | | |
Collapse
|
41
|
Naphade S, Embusch A, Madushani KL, Ring KL, Ellerby LM. Altered Expression of Matrix Metalloproteinases and Their Endogenous Inhibitors in a Human Isogenic Stem Cell Model of Huntington's Disease. Front Neurosci 2018; 11:736. [PMID: 29459817 PMCID: PMC5807396 DOI: 10.3389/fnins.2017.00736] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/18/2017] [Indexed: 11/23/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by a progressive movement disorder, psychiatric symptoms, and cognitive impairments. HD is caused by a CAG repeat expansion encoding a stretch of polyglutamine residues in the N-terminus of mutant huntingtin (mHTT) protein. Proteolytic processing of mHTT yields toxic fragments, which cause neurotoxicity and massive neuronal cell death predominantly in the striatum and cortex. Inhibition of mHTT cleavage reduces neuronal toxicity suggesting mHTT proteolysis contributes to HD pathogenesis. A previously conducted unbiased siRNA screen in our lab for known human proteases identified matrix metalloproteinases (MMPs) as modifiers of mHTT proteolysis and toxicity. To further study MMP activation in HD, isogenic HD, and control corrected (C116) neural stem cells (NSCs) prepared from HD patient-derived induced pluripotent stem cells were used to examine the role of MMPs and their endogenous inhibitors in this highly relevant model system. We found altered expression of MMP-2 and MMP-9 (gelatinases), MMP-3/10, and MMP-14, activity in HD-NSCs when compared to control C116-NSCs. Dysregulation in MMP activity was accompanied with concomitant changes in levels of endogenous inhibitors of MMPs, called tissue inhibitors of matrix metalloproteinases (TIMPs). Specifically, we observed decreased levels of TIMP-1 and TIMP-2 in HD-NSCs, suggesting part of the altered expression and activity of MMPs is due to lower abundance of these endogenous inhibitors. Immunofluorescence analysis revealed increased MMP/TIMP localization in the nucleus or aggregates of HD-NSCs, suggesting potential interaction with mHTT. TIMP-1 was found to associate with mHTT aggregates in discrete punctate structures in HD-NSCs. These events collectively contribute to increased neurotoxicity in HD. Previous characterization of these NSCs revealed transforming growth factor beta (TGF-β) pathway as the top dysregulated pathway in HD. TGF-β was significantly upregulated in HD-NSCs and addition of TGF-β to HD-NSCs was found to be neuroprotective. To determine if TGF-β regulated MMP and TIMP activity, C116- and HD-NSCs were exogenously treated with recombinant TGF-β. TIMP-1 levels were found to be elevated in response to TGF-β treatment, representing a potential mechanism through which elevated TGF-β levels confer neuroprotection in HD. Studying the mechanism of action of MMPs and TIMPs, and their interactions with mHTT in human isogenic patient-derived NSCs elucidates new mechanisms of HD neurotoxicity and will likely provide novel therapeutics for treatment of HD.
Collapse
Affiliation(s)
- Swati Naphade
- The Buck Institute for Research on Aging, Novato, CA, United States
| | | | | | - Karen L Ring
- The Buck Institute for Research on Aging, Novato, CA, United States.,California Institute of Regenerative Medicine, San Francisco, CA, United States
| | - Lisa M Ellerby
- The Buck Institute for Research on Aging, Novato, CA, United States
| |
Collapse
|
42
|
Brun MJ, Gomez EJ, Suh J. Stimulus-responsive viral vectors for controlled delivery of therapeutics. J Control Release 2017; 267:80-89. [PMID: 28842318 PMCID: PMC5723212 DOI: 10.1016/j.jconrel.2017.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/17/2017] [Accepted: 08/19/2017] [Indexed: 12/31/2022]
Abstract
Virus-based therapies have gained momentum as the next generation of treatments for a variety of serious diseases. In order to make these therapies more controllable, stimulus-responsive viral vectors capable of sensing and responding to specific environmental inputs are currently being developed. A number of viruses naturally respond to endogenous stimuli, such as pH, redox, and proteases, which are present at different concentrations in diseases and at different organ and organelle sites. Additionally, rather than relying on natural viral properties, efforts are underway to engineer viruses to respond to endogenous stimuli in new ways as well as to exogenous stimuli, such as temperature, magnetic field, and optical light. Viruses with stimulus-responsive capabilities, either nature-evolved or human-engineered, will be reviewed to capture the current state of the field. Stimulus-responsive viral vector design considerations as well as gaps in current research efforts will be identified.
Collapse
Affiliation(s)
- Mitchell J Brun
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, United States
| | - Eric J Gomez
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, TX, United States; Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, United States.
| |
Collapse
|
43
|
Pampalakis G, Sykioti VS, Ximerakis M, Stefanakou-Kalakou I, Melki R, Vekrellis K, Sotiropoulou G. KLK6 proteolysis is implicated in the turnover and uptake of extracellular alpha-synuclein species. Oncotarget 2017; 8:14502-14515. [PMID: 27845893 PMCID: PMC5362421 DOI: 10.18632/oncotarget.13264] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023] Open
Abstract
KLK6 is a serine protease highly expressed in the nervous system. In synucleinopathies, including Parkinson disease, the levels of KLK6 inversely correlate with α-synuclein in CSF. Recently, we suggested that recombinant KLK6 mediates the degradation of extracellular α-synuclein directly and via a proteolytic cascade that involves unidentified metalloproteinase(s). Here, we show that recombinant and naturally secreted KLK6 can readily cleave α-synuclein fibrils that have the potential for cell-to-cell propagation in “a prion-like mechanism”. Importantly, KLK6-deficient primary cortical neurons have increased ability for α-synuclein fibril uptake. We also demonstrate that KLK6 activates proMMP2, which in turn can cleave α-synuclein. The repertoire of proteases activated by KLK6 in a neuronal environment was analyzed by degradomic profiling, which also identified ADAMTS19 and showed that KLK6 has a limited number of substrates indicating specific biological functions such as the regulation of α-synuclein turnover. We generated adenoviral vectors for KLK6 delivery and demonstrated that the levels of extracellular α-synuclein can be reduced by neuronally secreted KLK6. Our findings open the possibility to exploit KLK6 as a novel therapeutic target for Parkinson disease and other synucleinopathies.
Collapse
Affiliation(s)
- Georgios Pampalakis
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Athens, Greece
| | - Vasia-Samantha Sykioti
- Center for Neurosciences, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Methodios Ximerakis
- Center for Neurosciences, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ioanna Stefanakou-Kalakou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Athens, Greece
| | - Ronald Melki
- Paris-Saclay Institute of Neuroscience, Centre National de la Recherche Scientifique, Gif-Sur-Yvette, France
| | - Kostas Vekrellis
- Center for Neurosciences, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Athens, Greece.,Center for Neurosciences, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
44
|
Rahimi S, Sayad A, Moslemi E, Ghafouri-Fard S, Taheri M. Blood assessment of the expression levels of matrix metalloproteinase 9 (MMP9) and its natural inhibitor, TIMP1 genes in Iranian schizophrenic patients. Metab Brain Dis 2017; 32:1537-1542. [PMID: 28578515 DOI: 10.1007/s11011-017-0043-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
Abstract
Schizophrenia (SCZ) is the most severe chronic mental disorder characterized by abnormal social behavior and disrupted emotions and thought. Like other complex neuropsychological disease, SCZ is caused by a combination of genetic and environmental factors but with a high concordance rate. So far, different genetic factors are revealed to be associated with increased risk of developing SCZ. One of the best ways to investigate the genetic basis of the complex disease is to discover the genetic underlying mechanisms of the defective clinical aspects of the patients. In this regard, genes involved in the developmental mechanisms of the brain such as long-term potentiation (LTP) process that is the basis of synaptic plasticity, memory and learning are considered as strong candidates for SCZ. The aim of the present study was to evaluate the expression levels of two genes that are involved in the LTP regulation in the developing and adult brain, Matrix metallopeptidase9 (MMP9) and TIMP metallopeptidase inhibitor 1 (TIMP1) genes in a blood assessment of schizophrenic patients in comparison to healthy controls by means of quantitative real time PCR. The results of the study showed a significant difference in MMP9/TIPM1 ratio between SCZ patients and healthy controls (P = 0.01). However, no significant difference was detected in the expression level of individual MMP9 and TIMP1 genes in SCZ patients versus healthy controls either in total numbers of subject or in sex based subgroups. Considering the relatively small sample size of the current study, there is a need to replicate this study with further investigations about the mechanism of association of these genes and their functions in the pathogenesis of the SCZ.
Collapse
Affiliation(s)
- Shahrzad Rahimi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran.
| | - Elham Moslemi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran
| | - Mohammad Taheri
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 1985717443, Tehran, Iran.
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Zhang D, Yang S, Toledo EM, Gyllborg D, Saltó C, Carlos Villaescusa J, Arenas E. Niche-derived laminin-511 promotes midbrain dopaminergic neuron survival and differentiation through YAP. Sci Signal 2017; 10:10/493/eaal4165. [DOI: 10.1126/scisignal.aal4165] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
46
|
Babula D, Kocot J, Horecka A, Baran M, Kurzepa J. Different patterns of gelatinolytic activity in pituitary macro- and microadenomas. Clin Neurol Neurosurg 2017; 158:90-92. [PMID: 28500926 DOI: 10.1016/j.clineuro.2017.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 03/28/2017] [Accepted: 05/03/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Gelatinases, Matrix MetalloProteinase(MMP)-2 and MMP-9, belong to zinc-dependent endopeptidases involved in several physiological and pathological processes including inflammation and tumor development. Because the information about the involvement of gelatinases in pituitary adenoma (PA) development are scant, our objective was the analysis of MMP-2 and MMP-9 activity in serum and tumor tissue of PA patients. PATIENTS AND METHODS Twenty one patients with PA (macroadenoma n=18, microadenoma n=3), qualified to the endoscopic resection of tumors were enrolled. Venous blood samples were collected before the surgery and PA tissue was collected during the surgery. Tissue material was homogenized in a buffer containing 0.1M Tris-HCl pH 7.4 and centrifuged. The supernatant was set to the equal protein content 18μg/sample. Protein level in tissue samples was estimated with Bradford method. MMP-2 and MMP-9 analysis in serum and tissue was performed with gelatin zymography. RESULTS The proteolytically activated forms of MMPs were not observed in the analyzed sera. Serum activities of MMP-2 and MMP-9 did not statistically differ between patients with micro and macroadenomas. The analysis of material obtained from tissue of microadenomas showed slightly lower activities of both forms of MMP-9 (pro-MMP-9 and MMP-9/lipokalin heterodimer). Simultaneously the increased activity of pro-MMP-2 in comparison to macroadenomas was observed. Although differences observed did not reach statistical significance, only in the case of microadenomas the presence of the active form of MMP-2 (molecular weight 65kDa band) was observed. CONCLUSION In the course of PA growth the change the biochemical profile of the gelatinolytic activity within the tumor tissue is observed. Initially, the higher activity of MMP-2 in microadenomas and elevated activity of MMP-9 in macroadenomas were detected.
Collapse
Affiliation(s)
- Daniel Babula
- Department of Neurosurgery and Neurotraumatology, Saint Edvige Provincial Hospital No. 2 in Rzeszow, Lwowska 60, 35-301 Rzeszów, PL, Poland
| | - Joanna Kocot
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4, 20-093 Lublin, PL, Poland
| | - Anna Horecka
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4, 20-093 Lublin, PL, Poland
| | - Marcin Baran
- Department of Maxillofacial Surgery, Medical University of Lublin, Poland
| | - Jacek Kurzepa
- Department of Medical Chemistry, Medical University of Lublin, Chodźki 4, 20-093 Lublin, PL, Poland.
| |
Collapse
|
47
|
Whole-exome sequencing of 228 patients with sporadic Parkinson's disease. Sci Rep 2017; 7:41188. [PMID: 28117402 PMCID: PMC5259721 DOI: 10.1038/srep41188] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, affecting 1% of the population over 65 years characterized clinically by both motor and non-motor symptoms accompanied by the preferential loss of dopamine neurons in the substantia nigra pars compacta. Here, we sequenced the exomes of 244 Parkinson's patients selected from the Oxford Parkinson's Disease Centre Discovery Cohort and, after quality control, 228 exomes were available for analyses. The PD patient exomes were compared to 884 control exomes selected from the UK10K datasets. No single non-synonymous (NS) single nucleotide variant (SNV) nor any gene carrying a higher burden of NS SNVs was significantly associated with PD status after multiple-testing correction. However, significant enrichments of genes whose proteins have roles in the extracellular matrix were amongst the top 300 genes with the most significantly associated NS SNVs, while regions associated with PD by a recent Genome Wide Association (GWA) study were enriched in genes containing PD-associated NS SNVs. By examining genes within GWA regions possessing rare PD-associated SNVs, we identified RAD51B. The protein-product of RAD51B interacts with that of its paralogue RAD51, which is associated with congenital mirror movements phenotypes, a phenotype also comorbid with PD.
Collapse
|
48
|
Oh SH, Kim HN, Park HJ, Shin JY, Kim DY, Lee PH. The Cleavage Effect of Mesenchymal Stem Cell and Its Derived Matrix Metalloproteinase-2 on Extracellular α-Synuclein Aggregates in Parkinsonian Models. Stem Cells Transl Med 2016; 6:949-961. [PMID: 28297586 PMCID: PMC5442774 DOI: 10.5966/sctm.2016-0111] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
Ample evidence has suggested that extracellular α‐synuclein aggregates would play key roles in the pathogenesis and progression of Parkinsonian disorders (PDs). In the present study, we investigated whether mesenchymal stem cells (MSCs) and their derived soluble factors could exert neuroprotective effects via proteolysis of extracellular α‐synuclein. When preformed α‐synuclein aggregates were incubated with MSC‐conditioned medium, α‐synuclein aggregates were disassembled, and insoluble and oligomeric forms of α‐synuclein were markedly decreased, thus leading to a significant increase in neuronal viability. In an animal study, MSC or MSC‐conditioned medium treatment decreased the expression of α‐synuclein oligomers and the induction of pathogenic α‐synuclein with an attenuation of apoptotic cell death signaling. Furthermore, we identified that matrix metalloproteinase‐2 (MMP‐2), a soluble factor derived from MSCs, played an important role in the degradation of extracellular α‐synuclein. Our data demonstrated that MSCs and their derived MMP‐2 exert neuroprotective properties through proteolysis of aggregated α‐synuclein in PD‐related microenvironments. Stem Cells Translational Medicine2017;6:949–961
Collapse
Affiliation(s)
- Se Hee Oh
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ha Na Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Jin Young Shin
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Yeol Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
49
|
Gentile E, Liuzzi GM. Marine pharmacology: therapeutic targeting of matrix metalloproteinases in neuroinflammation. Drug Discov Today 2016; 22:299-313. [PMID: 27697495 DOI: 10.1016/j.drudis.2016.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/18/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023]
Abstract
Alterations in matrix metalloproteinase (MMP) expression and activity are recognized as key pathogenetic events in several neurological disorders. This evidence makes MMPs possible therapeutic targets. The search for substances that can inhibit MMPs is moving progressively toward the screening of natural products. In particular, marine bioprospecting could be promising for the discovery of marine natural products with anti-MMP activities. Despite recent advances in this field, the possibility of using marine MMP inhibitors (MMPIs) for the treatment of neuroinflammation is still under-investigated. Here, we review the latest findings in this promising research field and the potential that marine MMPIs can have in the management and treatment of various neurological diseases.
Collapse
Affiliation(s)
- Eugenia Gentile
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Grazia M Liuzzi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy.
| |
Collapse
|
50
|
Le W, Wu J, Tang Y. Protective Microglia and Their Regulation in Parkinson's Disease. Front Mol Neurosci 2016; 9:89. [PMID: 27708561 PMCID: PMC5030290 DOI: 10.3389/fnmol.2016.00089] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/07/2016] [Indexed: 01/09/2023] Open
Abstract
Microglia-mediated neuroinflammation is a hallmark of Parkinson's disease (PD). In the brains of patients with PD, microglia have both neurotoxic and neuroprotective effects, depending on their activation state. In this review, we focus on recent research demonstrating the neuroprotective role of microglia in PD. Accumulating evidence indicates that the protective mechanisms of microglia may result from their regulation of transrepression pathways via nuclear receptors, anti-inflammatory responses, neuron-microglia crosstalk, histone modification, and microRNA regulation. All of these mechanisms work together to suppress the production of neurotoxic inflammatory components. However, during the progression of PD, the detrimental effects of inflammation overpower the protective actions of microglia. Therefore, an in-depth exploration of the mechanisms underlying microglial neuroprotection, and a means of promoting the transformation of microglia to the protective phenotype, are urgently needed for the treatment of PD.
Collapse
Affiliation(s)
- Weidong Le
- Center for Clinical Research on Neurological Diseases, First Affiliated Hospital, Dalian Medical University, DalianChina
| | - Junjiao Wu
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, ChangshaChina
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TXUSA
| | - Yu Tang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TXUSA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TXUSA
| |
Collapse
|