1
|
De S, Banerjee S, Rakshit P, Banerjee S, Kumar SKA. Unraveling the Ties: Type 2 Diabetes and Parkinson's Disease - A Nano-Based Targeted Drug Delivery Approach. Curr Diabetes Rev 2025; 21:32-58. [PMID: 38747222 DOI: 10.2174/0115733998291968240429111357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 02/26/2025]
Abstract
The link between Type 2 Diabetes (T2DM) and Parkinson's Disease (PD) dates back to the early 1960s, and ongoing research is exploring this association. PD is linked to dysregulation of dopaminergic pathways, neuroinflammation, decreased PPAR-γ coactivator 1-α, increased phosphoprotein enriched in diabetes, and accelerated α-Syn amyloid fibril production caused by T2DM. This study aims to comprehensively evaluate the T2DM-PD association and risk factors for PD in T2DM individuals. The study reviews existing literature using reputable sources like Scopus, ScienceDirect, and PubMed, revealing a significant association between T2DM and worsened PD symptoms. Genetic profiles of T2DM-PD individuals show similarities, and potential risk factors include insulin-resistance and dysbiosis of the gut-brain microbiome. Anti-diabetic drugs exhibit neuroprotective effects in PD, and nanoscale delivery systems like exosomes, micelles, and liposomes show promise in enhancing drug efficacy by crossing the Blood-Brain Barrier (BBB). Brain targeting for PD uses exosomes, micelles, liposomes, dendrimers, solid lipid nanoparticles, nano-sized polymers, and niosomes to improve medication and gene therapy efficacy. Surface modification of nanocarriers with bioactive compounds (such as angiopep, lactoferrin, and OX26) enhances α-Syn conjugation and BBB permeability. Natural exosomes, though limited, hold potential for investigating DM-PD pathways in clinical research. The study delves into the underlying mechanisms of T2DM and PD and explores current therapeutic approaches in the field of nano-based targeted drug delivery. Emphasis is placed on resolved and ongoing issues in understanding and managing both conditions.
Collapse
Affiliation(s)
- Sourav De
- Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Kolkata, 700126, West Bengal, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - Pallabita Rakshit
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - S K Ashok Kumar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
2
|
Ellis CPS, Tero BW, Potts CM, Malka KT, Yang X, Hamilton J, Vary C, Khalil A, Liaw L. Cellular Characteristics and Protein Signatures of Human Adipose Tissues from Donors With or Without Advanced Coronary Artery Disease. Biomedicines 2024; 12:2453. [PMID: 39595019 PMCID: PMC11592159 DOI: 10.3390/biomedicines12112453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Perivascular adipose tissue (PVAT) exerts a paracrine effect on blood vessels and our objective was to understand PVAT molecular signatures related to cardiovascular disease. Methods: We studied two groups: those undergoing mitral valve repair/replacement (VR, n = 16) and coronary artery bypass graft (CABG, n = 38). VR donors did not have coronary artery disease, whereas CABG donors had advanced coronary artery disease. Clinical and tissue pathologies and proteomics from adipose tissue were assessed. Results: Donors undergoing VR had a lower body mass index (p = 0.01), HbA1C (p = 0.0023), and incidence of diabetes (p = 0.022) compared to CABG. VR donors were overall healthier, with higher cardiac function compared to CABG donors, based on ejection fraction. Although adipose histopathology between groups was not markedly different, PVAT had smaller and more adipocytes compared to subcutaneous adipose tissues. These differences were validated by whole specimen automated morphological analysis, and anisotropy analysis showed small (2.8-7.5 μm) and large (22.8-64.4 μm) scale differences between perivascular and subcutaneous adipose tissue from CABG donors, and small scale changes (2.8-7.5 μm) between perivascular and subcutaneous adipose tissue from VR donors. Distinct protein signatures in PVAT and subcutaneous adipose tissue include those involved in secretion, exosomes and vesicles, insulin resistance, and adipocyte identity. Comparing PVAT and subcutaneous adipose tissue from CABG donors, there were 82 significantly different proteins identified with log fold change ≥ 0.3 or ≤-0.3 (p < 0.05). Using this threshold, there were 36 differences when comparing PVAT and subcutaneous adipose tissue from VR donors, 58 differences when comparing PVAT from CABG or VR donors, and 55 when comparing subcutaneous adipose tissue from CABG vs. VR donors. Conclusions: Routine histopathology cannot differentiate between PVAT from donors with or without coronary artery disease, but multiscale anisotropy analysis discriminated between these populations. Our mass spectrometry analysis identified a cohort of proteins that distinguish between adipose depots, and are also associated with the presence or absence of coronary artery disease.
Collapse
Affiliation(s)
- Caitlin P. S. Ellis
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Benjamin W. Tero
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Christian M. Potts
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Kimberly T. Malka
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Xuehui Yang
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
| | - Joshua Hamilton
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Calvin Vary
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| | - Andre Khalil
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
- CompuMAINE Lab, Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME 04469, USA
| | - Lucy Liaw
- MaineHealth Institute for Research, Scarborough, ME 04074, USA; (C.P.S.E.); (B.W.T.); (C.M.P.); (K.T.M.); (X.Y.); (C.V.)
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA; (J.H.); (A.K.)
| |
Collapse
|
3
|
Piedade de Souza T, Santana de Araújo G, Magalhães L, Cavalcante GC, Ribeiro-Dos-Santos A, Sena-Dos-Santos C, Silva CS, Eufraseo GL, de Freitas Escudeiro A, Soares-Souza GB, Santos-Lobato BL, Ribeiro-Dos-Santos Â. Unveiling differential gene co-expression networks and its effects on levodopa-induced dyskinesia. iScience 2024; 27:110835. [PMID: 39297167 PMCID: PMC11409023 DOI: 10.1016/j.isci.2024.110835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/25/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Levodopa-induced dyskinesia (LID) refers to involuntary motor movements of chronic use of levodopa in Parkinson's disease (PD) that negatively impact the overall well-being of people with this disease. The molecular mechanisms involved in LID were investigated through whole-blood transcriptomic analysis for differential gene expression and identification of new co-expression and differential co-expression networks. We found six differentially expressed genes in patients with LID, and 13 in patients without LID. We also identified 12 co-expressed genes exclusive to LID, and six exclusive hub genes involved in 23 gene-gene interactions in patients with LID. Convergently, we identified novel genes associated with PD and LID that play roles in mitochondrial dysfunction, dysregulation of lipid metabolism, and neuroinflammation. We observed significant changes in disease progression, consistent with previous findings of maladaptive plastic changes in the basal ganglia leading to the development of LID, including a chronic pro-inflammatory state in the brain.
Collapse
Affiliation(s)
- Tatiane Piedade de Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | | | | | - Giovanna C Cavalcante
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Arthur Ribeiro-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Camille Sena-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Caio Santos Silva
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Gracivane Lopes Eufraseo
- Laboratório de Neurologia Experimental, Universidade Federal do Pará, Belém 66073-000, Pará, Brazil
| | | | - Giordano Bruno Soares-Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
- Instituto Tecnológico Vale, Belém 66055-090, Pará, Brazil
| | | | - Ândrea Ribeiro-Dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará (UFPA), Belém 66073-005, Pará, Brazil
| |
Collapse
|
4
|
Shen J, Wang X, Wang M, Zhang H. Potential molecular mechanism of exercise reversing insulin resistance and improving neurodegenerative diseases. Front Physiol 2024; 15:1337442. [PMID: 38818523 PMCID: PMC11137309 DOI: 10.3389/fphys.2024.1337442] [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: 11/13/2023] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Neurodegenerative diseases are debilitating nervous system disorders attributed to various conditions such as body aging, gene mutations, genetic factors, and immune system disorders. Prominent neurodegenerative diseases include Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis. Insulin resistance refers to the inability of the peripheral and central tissues of the body to respond to insulin and effectively regulate blood sugar levels. Insulin resistance has been observed in various neurodegenerative diseases and has been suggested to induce the occurrence, development, and exacerbation of neurodegenerative diseases. Furthermore, an increasing number of studies have suggested that reversing insulin resistance may be a critical intervention for the treatment of neurodegenerative diseases. Among the numerous measures available to improve insulin sensitivity, exercise is a widely accepted strategy due to its convenience, affordability, and significant impact on increasing insulin sensitivity. This review examines the association between neurodegenerative diseases and insulin resistance and highlights the molecular mechanisms by which exercise can reverse insulin resistance under these conditions. The focus was on regulating insulin resistance through exercise and providing practical ideas and suggestions for future research focused on exercise-induced insulin sensitivity in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiawen Shen
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Xianping Wang
- School of Medicine, Taizhou University, Taizhou, China
| | - Minghui Wang
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Hu Zhang
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| |
Collapse
|
5
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Santos-García D, Martínez-Valbuena I, Agúndez JAG. Alpha-Synuclein in Peripheral Tissues as a Possible Marker for Neurological Diseases and Other Medical Conditions. Biomolecules 2023; 13:1263. [PMID: 37627328 PMCID: PMC10452242 DOI: 10.3390/biom13081263] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The possible usefulness of alpha-synuclein (aSyn) determinations in peripheral tissues (blood cells, salivary gland biopsies, olfactory mucosa, digestive tract, skin) and in biological fluids, except for cerebrospinal fluid (serum, plasma, saliva, feces, urine), as a marker of several diseases, has been the subject of numerous publications. This narrative review summarizes data from studies trying to determine the role of total, oligomeric, and phosphorylated aSyn determinations as a marker of various diseases, especially PD and other alpha-synucleinopathies. In summary, the results of studies addressing the determinations of aSyn in its different forms in peripheral tissues (especially in platelets, skin, and digestive tract, but also salivary glands and olfactory mucosa), in combination with other potential biomarkers, could be a useful tool to discriminate PD from controls and from other causes of parkinsonisms, including synucleinopathies.
Collapse
Affiliation(s)
| | | | - Elena García-Martín
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - Diego Santos-García
- Department of Neurology, CHUAC—Complejo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain;
| | - Iván Martínez-Valbuena
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 2S8, Canada;
| | - José A. G. Agúndez
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
6
|
Sabari SS, Balasubramani K, Iyer M, Sureshbabu HW, Venkatesan D, Gopalakrishnan AV, Narayanaswamy A, Senthil Kumar N, Vellingiri B. Type 2 Diabetes (T2DM) and Parkinson's Disease (PD): a Mechanistic Approach. Mol Neurobiol 2023:10.1007/s12035-023-03359-y. [PMID: 37118323 PMCID: PMC10144908 DOI: 10.1007/s12035-023-03359-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
Growing evidence suggest that there is a connection between Parkinson's disease (PD) and insulin dysregulation in the brain, whilst the connection between PD and type 2 diabetes mellitus (T2DM) is still up for debate. Insulin is widely recognised to play a crucial role in neuronal survival and brain function; any changes in insulin metabolism and signalling in the central nervous system (CNS) can lead to the development of various brain disorders. There is accumulating evidence linking T2DM to PD and other neurodegenerative diseases. In fact, they have a lot in common patho-physiologically, including insulin dysregulation, oxidative stress resulting in mitochondrial dysfunction, microglial activation, and inflammation. As a result, initial research should focus on the role of insulin and its molecular mechanism in order to develop therapeutic outcomes. In this current review, we will look into the link between T2DM and PD, the function of insulin in the brain, and studies related to impact of insulin in causing T2DM and PD. Further, we have also highlighted the role of various insulin signalling pathway in both T2DM and PD. We have also suggested that T2DM-targeting pharmacological strategies as potential therapeutic approach for individuals with cognitive impairment, and we have demonstrated the effectiveness of T2DM-prescribed drugs through current PD treatment trials. In conclusion, this investigation would fill a research gap in T2DM-associated Parkinson's disease (PD) with a potential therapy option.
Collapse
Affiliation(s)
- S Sri Sabari
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Kiruthika Balasubramani
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to Be University), Coimbatore, 641021, Tamil Nadu, India
| | - Harysh Winster Sureshbabu
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014, India
| | - Arul Narayanaswamy
- Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Nachimuthu Senthil Kumar
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796004, Mizoram, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India.
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
7
|
Wang Y, Bergström J, Ingelsson M, Westermark GT. Studies on alpha-synuclein and islet amyloid polypeptide interaction. Front Mol Biosci 2023; 10:1080112. [PMID: 36793785 PMCID: PMC9922763 DOI: 10.3389/fmolb.2023.1080112] [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: 10/25/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Introduction: Parkinson's disease and type 2 diabetes have both elements of local amyloid depositions in their pathogenesis. In Parkinson's disease, alpha-synuclein (aSyn) forms insoluble Lewy bodies and Lewy neurites in brain neurons, and in type 2 diabetes, islet amyloid polypeptide (IAPP) comprises the amyloid in the islets of Langerhans. In this study, we assessed the interaction between aSyn and IAPP in human pancreatic tissues, both ex vivo and in vitro. Material and Methods: The antibody-based detection techniques, proximity ligation assay (PLA), and immuno-TEM were used for co-localization studies. Bifluorescence complementation (BiFC) was used for interaction studies between IAPP and aSyn in HEK 293 cells. The Thioflavin T assay was used for studies of cross-seeding between IAPP and aSyn. ASyn was downregulated with siRNA, and insulin secretion was monitored using TIRF microscopy. Results: We demonstrate intracellular co-localization of aSyn with IAPP, while aSyn is absent in the extracellular amyloid deposits. ASyn reactivity is present in the secretory granules of β-cells and some α-cells in human islets. The BiFC-expression of aSyn/aSyn and IAPP/IAPP in HEK293 cells resulted in 29.3% and 19.7% fluorescent cells, respectively, while aSyn/IAPP co-expression resulted in ∼10% fluorescent cells. Preformed aSyn fibrils seeded IAPP fibril formation in vitro, but adding preformed IAPP seeds to aSyn did not change aSyn fibrillation. In addition, mixing monomeric aSyn with monomeric IAPP did not affect IAPP fibril formation. Finally, the knockdown of endogenous aSyn did not affect β cell function or viability, nor did overexpression of aSyn affect β cell viability. Discussion: Despite the proximity of aSyn and IAPP in β-cells and the detected capacity of preformed aSyn fibrils to seed IAPP in vitro, it is still an open question if an interaction between the two molecules is of pathogenic significance for type 2 diabetes.
Collapse
Affiliation(s)
- Ye Wang
- Departments of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Departments ofPublic Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Departments ofPublic Health and Caring Sciences, Uppsala University, Uppsala, Sweden,Krembil Brain Institute, University Health Network, Toronto, ON, Canada,Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Gunilla T. Westermark
- Departments of Medical Cell Biology, Uppsala University, Uppsala, Sweden,*Correspondence: Gunilla T. Westermark,
| |
Collapse
|
8
|
Ebrahimzadeh Peer M, Fallahmohammadi Z, Akbari A. The effect of progressive endurance training and extract of black winter truffle on proteins levels and expression of hippocampus α-synuclein and HSF1 in the healthy and diabetic rats. Metabol Open 2023; 17:100232. [PMID: 36785616 PMCID: PMC9918783 DOI: 10.1016/j.metop.2023.100232] [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: 12/05/2022] [Revised: 01/09/2023] [Accepted: 01/22/2023] [Indexed: 01/29/2023] Open
Abstract
Aim The research aimed to investigate the effect of endurance running and T. Brumale extract on α-Syn and HSF1 in the brain and serum of healthy and diabetic rats. Methods A total of 40 Wistar rats were randomly divided into eight groups: Control (C), Exercise (E), Control-Tuber (T), Exercise-Tuber (ET), Control-Diabetes (D), Exercise-Diabetes (ED), Control-Diabetes-Tuber (CDT), and Exercise-Diabetes-Tuber (EDT). The endurance running was carried out five times per week for five weeks. The hippocampus and the serum α-Syn and HSF1 were measured using an enzyme-linked immunosorbent assay method. Results The brain α-Syn levels were higher in diabetic groups than in the healthy groups, but insignificantly (P ≤ 0.05). The brain α-Syn level significantly increased in the EDT group compared to the T group (P ≤ 0.05). The serum level of α-Syn in the ED group was significantly higher than in the E and D groups (P ≤ 0.05). The brain HSF1 level was significantly higher in the ED group compared to the D group (P ≤ 0.05). The gene expression of hsf1 was significantly reduced in the E group compared to the other groups and the EDT group compared to ED and CDT groups (P ≤ 0.05). Furthermore, the serum HSF1 level significantly increased in the ED group compared to the D group (P ≤ 0.05). Conclusion The results of this study suggest that progressive endurance running may improve neuroprotective conditions in diabetic patients by increasing HSF1 in the brain.
Collapse
Affiliation(s)
- Mojtaba Ebrahimzadeh Peer
- Exercise Physiology Department, Sports Sciences Faculty, University of Mazandaran, Babolsar, Mazandaran Province, Iran
| | - Ziya Fallahmohammadi
- Exercise Physiology Department, Sports Sciences Faculty, University of Mazandaran, Babolsar, Mazandaran Province, Iran,Corresponding author
| | - Abolfazl Akbari
- Physiology Department, Veterinary Medicine School, University of Shiraz, Shiraz, Fars Province, Iran
| |
Collapse
|
9
|
Takami Y, Wang C, Nakagami H, Yamamoto K, Nozato Y, Imaizumi Y, Nagasawa M, Takeshita H, Nakajima T, Takeda S, Takeya Y, Kaneda Y, Rakugi H. Novel pathophysiological roles of α-synuclein in age-related vascular endothelial dysfunction. FASEB J 2022; 36:e22555. [PMID: 36125010 DOI: 10.1096/fj.202101621r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 11/11/2022]
Abstract
Although α-synuclein (SNCA) is a well-known pathological molecule involved in synucleinopathy in neurons, its physiological roles remain largely unknown. We reported that serum SNCA levels have a close inverse correlation with blood pressure and age, which indicates the involvement of SNCA in age-related endothelial dysfunction. Therefore, this study aimed to elucidate the molecular functions of SNCA in the endothelium. We confirmed that SNCA was expressed in and secreted from endothelial cells (ECs). Exogenous treatment with recombinant SNCA (rSNCA) activated the Akt-eNOS axis and increased nitric oxide production in ECs. Treatment with rSNCA also suppressed TNF-α- and palmitic acid-induced NF-κB activation, leading to the suppression of VCAM-1 upregulation and restoration of eNOS downregulation in ECs. As for endogenous SNCA expression, replicative senescence resulted in the attenuation of SNCA expression in cultured ECs, similar to the effects of physiological aging on mice aortas. The siRNA-mediated silencing of SNCA consistently resulted in senescent phenotypes, such as eNOS downregulation, increased β-gal activity, decreased Sirt1 expression, and increased p53 expression, in ECs. Ex vivo assessment of endothelial functions using aortic rings revealed impaired endothelium-dependent acetylcholine-induced relaxation in SNCA knockout (KO) mice. Furthermore, SNCA KO mice, especially those on a high-fat diet, displayed elevated blood pressure compared with wild-type mice; this could be eNOS dysfunction-dependent because of the lower difference caused by L-NAME administration. These results indicate that exogenous and endogenous SNCA in ECs might physiologically maintain vascular integrity, and age-related endothelial dysfunction might be partially ascribed to loss-of-function of SNCA in ECs.
Collapse
Affiliation(s)
- Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Cheng Wang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Imaizumi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motonori Nagasawa
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hikari Takeshita
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsuneo Nakajima
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka, Japan.,Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Yasushi Takeya
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
10
|
Metabolism and memory: α-synuclein level in children with obesity and children with type 1 diabetes; relation to glucotoxicity, lipotoxicity and executive functions. Int J Obes (Lond) 2022; 46:2040-2049. [PMID: 36153375 PMCID: PMC9584809 DOI: 10.1038/s41366-022-01222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/19/2022] [Accepted: 09/05/2022] [Indexed: 11/08/2022]
Abstract
Abstract
Background/Objectives
Children with obesity and those with type 1diabetes (T1D) exhibit subtle neurocognitive deficits, the mechanism of which remains unknown. α-synuclein plays a fundamental role in neurodegeneration. Moreover, its role in glucose and lipids metabolism is emerging. This study aims to assess whether α-synuclein is correlated with the degree of neurodegeneration in children with obesity and those with T1D in comparison to healthy controls and correlate it to various neurocognitive and metabolic parameters.
Subjects/Methods
Forty children with obesity, 40 children with T1D and 40 matched-healthy controls were assessed for anthropometric measurements and blood-pressure. Cognitive evaluation was performed using Stanford–Binet scale and Barkley Deficits in Executive Functioning (EF) Scale-Children and Adolescents. α-synuclein, fasting lipids and glucose were measured with calculation of the homeostatic model of insulin-resistance and estimated-glucose disposal rate.
Results
Children with obesity and those with T1D had significantly higher α-synuclein (p < 0.001) and total EF percentile (p = 0.001) than controls. α-synuclein was negatively correlated to total IQ (p < 0.001 and p = 0.001), and positively correlated with total EF percentile (p = 0.009 and p = 0.001) and EF symptom count percentile (p = 0.005 and p < 0.001) in children with T1D and obesity, respectively. Multivariate-regression revealed that α-synuclein was independently related to age (p = 0.028), diabetes-duration (p = 0.006), HbA1C% (p = 0.034), total IQ (p = 0.013) and EF symptom count percentile (p = 0.003) among children with T1D, and to diastolic blood-pressure percentile (p = 0.013), waist/hip ratio SDS (p = 0.007), total EF percentile (P = 0.033) and EF symptom count percentile (p < 0.001) in children with obesity.
Conclusion
α-synuclein could have a mechanistic role in neurocognitive deficit among children with obesity and T1D.
Collapse
|
11
|
De Iuliis A, Montinaro E, Fatati G, Plebani M, Colosimo C. Diabetes mellitus and Parkinson's disease: dangerous liaisons between insulin and dopamine. Neural Regen Res 2022; 17:523-533. [PMID: 34380882 PMCID: PMC8504381 DOI: 10.4103/1673-5374.320965] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 11/13/2022] Open
Abstract
The relationship between diabetes mellitus and Parkinson's disease has been described in several epidemiological studies over the 1960s to date. Molecular studies have shown the possible functional link between insulin and dopamine, as there is strong evidence demonstrating the action of dopamine in pancreatic islets, as well as the insulin effects on feeding and cognition through central nervous system mechanism, largely independent of glucose utilization. Therapies used for the treatment of type 2 diabetes mellitus appear to be promising candidates for symptomatic and/or disease-modifying action in neurodegenerative diseases including Parkinson's disease, while an old dopamine agonist, bromocriptine, has been repositioned for the type 2 diabetes mellitus treatment. This review will aim at reappraising the different studies that have highlighted the dangerous liaisons between diabetes mellitus and Parkinson's disease.
Collapse
Affiliation(s)
| | - Ennio Montinaro
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| | | | - Mario Plebani
- Department of Medicine-DiMED, University of Padova, Italy
- Department of Medicine-DiMED, University of Padova, Padova, Italy; Department of Laboratory Medicine-Hospital of Padova, Padova, Italy
| | - Carlo Colosimo
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| |
Collapse
|
12
|
Barba L, Paolini Paoletti F, Bellomo G, Gaetani L, Halbgebauer S, Oeckl P, Otto M, Parnetti L. Alpha and Beta Synucleins: From Pathophysiology to Clinical Application as Biomarkers. Mov Disord 2022; 37:669-683. [PMID: 35122299 PMCID: PMC9303453 DOI: 10.1002/mds.28941] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
The synuclein family includes three neuronal proteins, named α‐synuclein, β‐synuclein, and γ‐synuclein, that have peculiar structural features. α‐synuclein is largely known for being a key protein in the pathophysiology of Parkinson's disease (PD) and other synucleinopathies, namely, dementia with Lewy bodies and multisystem atrophy. The role of β‐synuclein and γ‐synuclein is less well understood in terms of physiological functions and potential contribution to human diseases. α‐synuclein has been investigated extensively in both cerebrospinal fluid (CSF) and blood as a potential biomarker for synucleinopathies. Recently, great attention has been also paid to β‐synuclein, whose CSF and blood levels seem to reflect synaptic damage and neurodegeneration independent of the presence of synucleinopathy. In this review, we aim to provide an overview on the pathophysiological roles of the synucleins. Because γ‐synuclein has been poorly investigated in the field of synucleinopathy and its pathophysiological roles are far from being clear, we focus on the interactions between α‐synuclein and β‐synuclein in PD. We also discuss the role of α‐synuclein and β‐synuclein as potential biomarkers to improve the diagnostic characterization of synucleinopathies, thus highlighting their potential application in clinical trials for disease‐modifying therapies. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Lorenzo Barba
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
- Department of Neurology University of Ulm Ulm Germany
- Department of Neurology Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | - Federico Paolini Paoletti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Giovanni Bellomo
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Lorenzo Gaetani
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| | | | - Patrick Oeckl
- Department of Neurology University of Ulm Ulm Germany
- German Center for Neurodegenerative Disorders Ulm (DZNE e. V.) Ulm Germany
| | - Markus Otto
- Department of Neurology University of Ulm Ulm Germany
- Department of Neurology Martin‐Luther‐University Halle‐Wittenberg Halle/Saale Germany
| | - Lucilla Parnetti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery University of Perugia Perugia Italy
| |
Collapse
|
13
|
Mahmoud AM. An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions. Int J Mol Sci 2022; 23:ijms23031341. [PMID: 35163268 PMCID: PMC8836029 DOI: 10.3390/ijms23031341] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely explain the massive growth in obesity over the last few decades. Accumulating evidence suggests that modern lifestyle characteristics such as the intake of energy-dense foods, adopting sedentary behavior, or exposure to environmental factors such as industrial endocrine disruptors all contribute to the rising obesity epidemic. Recent advances in the study of DNA and its alterations have considerably increased our understanding of the function of epigenetics in regulating energy metabolism and expenditure in obesity and metabolic diseases. These epigenetic modifications influence how DNA is transcribed without altering its sequence. They are dynamic, reflecting the interplay between the body and its surroundings. Notably, these epigenetic changes are reversible, making them appealing targets for therapeutic and corrective interventions. In this review, I discuss how these epigenetic modifications contribute to the disordered energy metabolism in obesity and to what degree lifestyle and weight reduction strategies and pharmacological drugs can restore energy balance by restoring normal epigenetic profiles.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
14
|
Lary CW, Rosen CJ, Kiel DP. Osteoporosis and Dementia: Establishing a Link. J Bone Miner Res 2021; 36:2103-2105. [PMID: 34515377 PMCID: PMC8595864 DOI: 10.1002/jbmr.4431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/29/2021] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Boutros SW, Raber J, Unni VK. Effects of Alpha-Synuclein Targeted Antisense Oligonucleotides on Lewy Body-Like Pathology and Behavioral Disturbances Induced by Injections of Pre-Formed Fibrils in the Mouse Motor Cortex. JOURNAL OF PARKINSONS DISEASE 2021; 11:1091-1115. [PMID: 34057097 PMCID: PMC8461707 DOI: 10.3233/jpd-212566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Alpha-synuclein (αsyn) characterizes neurodegenerative diseases known as synucleinopathies. The phosphorylated form (psyn) is the primary component of protein aggregates known as Lewy bodies (LBs), which are the hallmark of diseases such as Parkinson’s disease (PD). Synucleinopathies might spread in a prion-like fashion, leading to a progressive emergence of symptoms over time. αsyn pre-formed fibrils (PFFs) induce LB-like pathology in wild-type (WT) mice, but questions remain about their progressive spread and their associated effects on behavioral performance. Objective: To characterize the behavioral, cognitive, and pathological long-term effects of LB-like pathology induced after bilateral motor cortex PFF injection in WT mice and to assess the ability of mouse αsyn-targeted antisense oligonucleotides (ASOs) to ameliorate those effects. Methods: We induced LB-like pathology in the motor cortex and connected brain regions of male WT mice using PFFs. Three months post-PFF injection (mpi), we assessed behavioral and cognitive performance. We then delivered a targeted ASO via the ventricle and assessed behavioral and cognitive performance 5 weeks later, followed by pathological analysis. Results: At 3 and 6 mpi, PFF-injected mice showed mild, progressive behavioral deficits. The ASO reduced total αsyn and psyn protein levels, and LB-like pathology, but was also associated with some deleterious off-target effects not involving lowering of αsyn, such as a decline in body weight and impairments in motor function. Conclusions: These results increase understanding of the progressive nature of the PFF model and support the therapeutic potential of ASOs, though more investigation into effects of ASO-mediated reduction in αsyn on brain function is needed.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.,Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Departments of Psychiatry and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| | - Vivek K Unni
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Jungers Center for Neurosciences Research and OHSU Parkinson Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
16
|
Wijesekara N, Ahrens R, Wu L, Langman T, Tandon A, Fraser PE. α-Synuclein Regulates Peripheral Insulin Secretion and Glucose Transport. Front Aging Neurosci 2021; 13:665348. [PMID: 34393754 PMCID: PMC8361797 DOI: 10.3389/fnagi.2021.665348] [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: 02/07/2021] [Accepted: 07/05/2021] [Indexed: 11/23/2022] Open
Abstract
Aim Population based studies indicate a positive association between type 2 diabetes (T2D) and Parkinson’s disease (PD) where there is an increased risk of developing PD in patients with T2D. PD is characterized by the abnormal accumulation of intraneuronal aggregated α-synuclein (α-syn) in Lewy bodies, which negatively impact neuronal viability. α-syn is also expressed in both pancreatic islets and skeletal muscle, key players in glucose regulation. Therefore, we examined the functional role of α-syn in these tissues. Methods Using mice lacking, overexpressing or transiently injected with α-syn, effects on glucose and insulin tolerance and insulin secretion were determined, with further characterization of the effects on GLUT4 translocation using GLUT4myc myotubes. Results Mice genetically ablated for α-syn became glucose intolerant and insulin resistant with hyperinsulinemia and reduced glucose-stimulated insulin secretion (GSIS). Mice overexpressing human α-syn are more insulin senstive and glucose tolerant compared to controls with increased GSIS. Injection of purified α-syn monomers also led to improved glucose tolerance and insulin sensitivity with hightened GSIS. α-syn monomer treatments increased surface GLUT4 levels in myotubes but without any significant change in Akt phosphorylation. The increase in cell surface GLUT4 was largely due to a large reduction in GLUT4 endocytosis, however, with a compensatory reduction in GLUT4 exocytosis. Conclusion Cumulatively, this data suggests that α-syn modulates both pancreatic beta cell function and glucose transport in peripheral tissues, thereby playing a pivitol role in the maintenance of normal glucose homeostasis.
Collapse
Affiliation(s)
- Nadeeja Wijesekara
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Rosemary Ahrens
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ling Wu
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Tammy Langman
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Manzanza NDO, Sedlackova L, Kalaria RN. Alpha-Synuclein Post-translational Modifications: Implications for Pathogenesis of Lewy Body Disorders. Front Aging Neurosci 2021; 13:690293. [PMID: 34248606 PMCID: PMC8267936 DOI: 10.3389/fnagi.2021.690293] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Lewy Body Disorders (LBDs) lie within the spectrum of age-related neurodegenerative diseases now frequently categorized as the synucleinopathies. LBDs are considered to be among the second most common form of neurodegenerative dementias after Alzheimer's disease. They are progressive conditions with variable clinical symptoms embodied within specific cognitive and behavioral disorders. There are currently no effective treatments for LBDs. LBDs are histopathologically characterized by the presence of abnormal neuronal inclusions commonly known as Lewy Bodies (LBs) and extracellular Lewy Neurites (LNs). The inclusions predominantly comprise aggregates of alpha-synuclein (aSyn). It has been proposed that post-translational modifications (PTMs) such as aSyn phosphorylation, ubiquitination SUMOylation, Nitration, o-GlcNacylation, and Truncation play important roles in the formation of toxic forms of the protein, which consequently facilitates the formation of these inclusions. This review focuses on the role of different PTMs in aSyn in the pathogenesis of LBDs. We highlight how these PTMs interact with aSyn to promote misfolding and aggregation and interplay with cell membranes leading to the potential functional and pathogenic consequences detected so far, and their involvement in the development of LBDs.
Collapse
Affiliation(s)
- Nelson de Oliveira Manzanza
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lucia Sedlackova
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
18
|
Emerging Roles of Metallothioneins in Beta Cell Pathophysiology: Beyond and Above Metal Homeostasis and Antioxidant Response. BIOLOGY 2021; 10:biology10030176. [PMID: 33652748 PMCID: PMC7996892 DOI: 10.3390/biology10030176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Defective insulin secretion by pancreatic beta cells is key for the development of type 2 diabetes but the precise mechanisms involved are poorly understood. Metallothioneins are metal binding proteins whose precise biological roles have not been fully characterized. Available evidence indicated that Metallothioneins are protective cellular effectors involved in heavy metal detoxification, metal ion homeostasis and antioxidant defense. This concept has however been challenged by emerging evidence in different medical research fields revealing novel negative roles of Metallothioneins, including in the context of diabetes. In this review, we gather and analyze the available knowledge regarding the complex roles of Metallothioneins in pancreatic beta cell biology and insulin secretion. We comprehensively analyze the evidence showing positive effects of Metallothioneins on beta cell function and survival as well as the emerging evidence revealing negative effects and discuss the possible underlying mechanisms. We expose in parallel findings from other medical research fields and underscore unsettled questions. Then, we propose some future research directions to improve knowledge in the field. Abstract Metallothioneins (MTs) are low molecular weight, cysteine-rich, metal-binding proteins whose precise biological roles have not been fully characterized. Existing evidence implicated MTs in heavy metal detoxification, metal ion homeostasis and antioxidant defense. MTs were thus categorized as protective effectors that contribute to cellular homeostasis and survival. This view has, however, been challenged by emerging evidence in different medical fields revealing novel pathophysiological roles of MTs, including inflammatory bowel disease, neurodegenerative disorders, carcinogenesis and diabetes. In the present focused review, we discuss the evidence for the role of MTs in pancreatic beta-cell biology and insulin secretion. We highlight the pattern of specific isoforms of MT gene expression in rodents and human beta-cells. We then discuss the mechanisms involved in the regulation of MTs in islets under physiological and pathological conditions, particularly type 2 diabetes, and analyze the evidence revealing adaptive and negative roles of MTs in beta-cells and the potential mechanisms involved. Finally, we underscore the unsettled questions in the field and propose some future research directions.
Collapse
|
19
|
Odenkirk MT, Stratton KG, Gritsenko MA, Bramer LM, Webb-Robertson BJM, Bloodsworth KJ, Weitz KK, Lipton AK, Monroe ME, Ash JR, Fourches D, Taylor BD, Burnum-Johnson KE, Baker ES. Unveiling molecular signatures of preeclampsia and gestational diabetes mellitus with multi-omics and innovative cheminformatics visualization tools. Mol Omics 2020; 16:521-532. [PMID: 32966491 PMCID: PMC7736332 DOI: 10.1039/d0mo00074d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To fully enable the development of diagnostic tools and progressive pharmaceutical drugs, it is imperative to understand the molecular changes occurring before and during disease onset and progression. Systems biology assessments utilizing multi-omic analyses (e.g. the combination of proteomics, lipidomics, genomics, etc.) have shown enormous value in determining molecules prevalent in diseases and their associated mechanisms. Herein, we utilized multi-omic evaluations, multi-dimensional analysis methods, and new cheminformatics-based visualization tools to provide an in depth understanding of the molecular changes taking place in preeclampsia (PRE) and gestational diabetes mellitus (GDM) patients. Since PRE and GDM are two prevalent pregnancy complications that result in adverse health effects for both the mother and fetus during pregnancy and later in life, a better understanding of each is essential. The multi-omic evaluations performed here provide new insight into the end-stage molecular profiles of each disease, thereby supplying information potentially crucial for earlier diagnosis and treatments.
Collapse
Affiliation(s)
- Melanie T Odenkirk
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sportelli C, Urso D, Jenner P, Chaudhuri KR. Metformin as a Potential Neuroprotective Agent in Prodromal Parkinson's Disease-Viewpoint. Front Neurol 2020; 11:556. [PMID: 32595595 PMCID: PMC7304367 DOI: 10.3389/fneur.2020.00556] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022] Open
Abstract
To date, there are no clinically effective neuroprotective or disease-modifying treatments that can halt Parkinson's disease (PD) progression. The current clinical approach focuses on symptomatic management. This failure may relate to the complex neurobiology underpinning the development of PD and the absence of true translational animal models. In addition, clinical diagnosis of PD relies on presentation of motor symptoms which occur when the neuropathology is already established. These multiple factors could contribute to the unsuccessful development of neuroprotective treatments for PD. Prodromal symptoms develop years prior to formal diagnosis and may provide an excellent tool for early diagnosis and better trial design. Patients with idiopathic rapid eye movement behavior disorder (iRBD) have the highest risk of developing PD and could represent an excellent group to include in neuroprotective trials for PD. In addition, repurposing drugs with excellent safety profiles is an appealing strategy to accelerate drug discovery. The anti-diabetic drug metformin has been shown to target diverse cellular pathways implicated in PD progression. Multiple studies have, additionally, observed the benefits of metformin to counteract other age-related diseases. The purpose of this viewpoint is to discuss metformin's neuroprotective potential by outlining relevant mechanisms of action and the selection of iRBD patients for future clinical trials in PD.
Collapse
Affiliation(s)
- Carolina Sportelli
- National Parkinson Foundation International Centre of Excellence, King's College Hospital, London, United Kingdom
| | - Daniele Urso
- National Parkinson Foundation International Centre of Excellence, King's College Hospital, London, United Kingdom.,Institute of Psychiatry, Psychology & Neuroscience, King's College, London, United Kingdom
| | - Peter Jenner
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College, London, United Kingdom
| | - K Ray Chaudhuri
- National Parkinson Foundation International Centre of Excellence, King's College Hospital, London, United Kingdom.,Institute of Psychiatry, Psychology & Neuroscience, King's College, London, United Kingdom
| |
Collapse
|
21
|
Analysis of the Relationship between Type II Diabetes Mellitus and Parkinson's Disease: A Systematic Review. PARKINSONS DISEASE 2019; 2019:4951379. [PMID: 31871617 PMCID: PMC6906831 DOI: 10.1155/2019/4951379] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022]
Abstract
In the early sixties, a discussion started regarding the association between Parkinson's disease (PD) and type II diabetes mellitus (T2DM). Today, this potential relationship is still a matter of debate. This review aims to analyze both diseases concerning causal relationships and treatments. A total of 104 articles were found, and studies on animal and “in vitro” models showed that T2DM causes neurological alterations that may be associated with PD, such as deregulation of the dopaminergic system, a decrease in the expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), an increase in the expression of phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes 15 (PED/PEA-15), and neuroinflammation, as well as acceleration of the formation of alpha-synuclein amyloid fibrils. In addition, clinical studies described that Parkinson's symptoms were notably worse after the onset of T2DM, and seven deregulated genes were identified in the DNA of T2DM and PD patients. Regarding treatment, the action of antidiabetic drugs, especially incretin mimetic agents, seems to confer certain degree of neuroprotection to PD patients. In conclusion, the available evidence on the interaction between T2DM and PD justifies more robust clinical trials exploring this interaction especially the clinical management of patients with both conditions.
Collapse
|
22
|
Pei Y, Maitta RW. Alpha synuclein in hematopoiesis and immunity. Heliyon 2019; 5:e02590. [PMID: 31692680 PMCID: PMC6806402 DOI: 10.1016/j.heliyon.2019.e02590] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/13/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative condition and intracellular deposition of Lewy bodies in the substantia nigra (SN), which can cause dopaminergic neuronal death, is the hallmark of this syndrome. α-synuclein (syn) is a small protein expressed mainly in neurons but can also be found in a number of tissues. It can be present as a soluble monomer under normal physiological conditions, but can be toxic in its oligomeric or fibrillary forms. Most of the available literature has focused on the effects of α-syn pathology in the mechanisms leading to PD. However, the normal functions of α-syn still remain to be fully elucidated. Notably, α-syn in the hematopoietic system seems to mediate important functions as indicated by anemia and incomplete cell maturation when this protein is absent. This review will summarize basic genetic and structural findings, and critical information that suggests an essential role of α-syn in the development and activation of the hematopoietic system and immunity.
Collapse
Affiliation(s)
- Yu Pei
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Robert W. Maitta
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
23
|
Velázquez AM, Roversi K, Dillenburg-Pilla P, Rodrigues RF, Zárate-Bladés CR, Prediger RDS, Izídio GS. The influence of chromosome 4 on metabolism and spatial memory in SHR and SLA16 rat strains. Behav Brain Res 2019; 370:111966. [PMID: 31125622 DOI: 10.1016/j.bbr.2019.111966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/13/2019] [Accepted: 05/20/2019] [Indexed: 11/16/2022]
Abstract
The Spontaneously Hypertensive Rat (SHR) has been proposed as a good model to study the pathways related to neurodegenerative diseases and glucose intolerance. Our research group developed the SLA16 (SHR.LEW-Anxrr16) congenic strain, which is genetically identical to the SHR strain, except for a locus on chromosome 4 (DGR). We applied in silico analysis on DGR to evaluate the association of their genes with neurobiological and metabolic pathways. After, we characterized cholesterol, triglycerides, metabolism of glucose and the behavioral performance of young (2 months old) and adult (8 months old) SHR and SLA16 rats in the open field, object location and water maze tasks. Finally, naïve young rats were repeatedly treated with metformin (200 mg/kg; v.o.) and evaluated in the same tests. Bioinformatics analysis showed that DGR presents genes related to glucose metabolism, oxidative damage and neurodegenerative diseases. Young SLA16 presented higher cholesterol, triglycerides, glucose and locomotion in the open field than SHR rats. In adulthood, SLA16 rats presented high triglycerides and locomotion in the open field and impairment on spatial learning and memory. Finally, the treatment with metformin decreased the glucose tolerance curve and also improved long-term memory in SLA16 rats. These results indicate that DGR presents genes associated with metabolic pathways and neurobiological processes that may produce alterations in glucose metabolism and spatial learning/memory. Therefore, we suggest that SHR and SLA16 strains could be important for the study of genes and subsequent mechanisms that produce metabolic glucose alterations and age-related cognitive deficits.
Collapse
Affiliation(s)
- Ana Magdalena Velázquez
- Departamento de Biologia Celular, Embriologia e Genética, Universidade Federal de Santa Catarina, Florianópolis, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Katiane Roversi
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Patricia Dillenburg-Pilla
- Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | - Carlos R Zárate-Bladés
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Rui Daniel S Prediger
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Geison Souza Izídio
- Departamento de Biologia Celular, Embriologia e Genética, Universidade Federal de Santa Catarina, Florianópolis, Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil; Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
| |
Collapse
|
24
|
Fiory F, Perruolo G, Cimmino I, Cabaro S, Pignalosa FC, Miele C, Beguinot F, Formisano P, Oriente F. The Relevance of Insulin Action in the Dopaminergic System. Front Neurosci 2019; 13:868. [PMID: 31474827 PMCID: PMC6706784 DOI: 10.3389/fnins.2019.00868] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
The advances in medicine, together with lifestyle modifications, led to a rising life expectancy. Unfortunately, however, aging is accompanied by an alarming boost of age-associated chronic pathologies, including neurodegenerative and metabolic diseases. Interestingly, a non-negligible interplay between alterations of glucose homeostasis and brain dysfunction has clearly emerged. In particular, epidemiological studies have pointed out a possible association between Type 2 Diabetes (T2D) and Parkinson’s Disease (PD). Insulin resistance, one of the major hallmark for etiology of T2D, has a detrimental influence on PD, negatively affecting PD phenotype, accelerating its progression and worsening cognitive impairment. This review aims to provide an exhaustive analysis of the most recent evidences supporting the key role of insulin resistance in PD pathogenesis. It will focus on the relevance of insulin in the brain, working as pro-survival neurotrophic factor and as a master regulator of neuronal mitochondrial function and oxidative stress. Insulin action as a modulator of dopamine signaling and of alpha-synuclein degradation will be described in details, too. The intriguing idea that shared deregulated pathogenic pathways represent a link between PD and insulin resistance has clinical and therapeutic implications. Thus, ongoing studies about the promising healing potential of common antidiabetic drugs such as metformin, exenatide, DPP IV inhibitors, thiazolidinediones and bromocriptine, will be summarized and the rationale for their use to decelerate neurodegeneration will be critically assessed.
Collapse
Affiliation(s)
- Francesca Fiory
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesca Chiara Pignalosa
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT "Genomic of Diabetes," Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
25
|
Ugalde CL, Lawson VA, Finkelstein DI, Hill AF. The role of lipids in α-synuclein misfolding and neurotoxicity. J Biol Chem 2019; 294:9016-9028. [PMID: 31064841 DOI: 10.1074/jbc.rev119.007500] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The misfolding and aggregation of α-synuclein (αsyn) in the central nervous system is associated with a group of neurodegenerative disorders referred to as the synucleinopathies. In addition to being a pathological hallmark of disease, it is now well-established that upon misfolding, αsyn acquires pathogenic properties, such as neurotoxicity, that can contribute to disease development. The mechanisms that produce αsyn misfolding and the molecular events underlying the neuronal damage caused by these misfolded species are not well-defined. A consistent observation that may be relevant to αsyn's pathogenicity is its ability to associate with lipids. This appears important not only to how αsyn aggregates, but also to the mechanism by which the misfolded protein causes intracellular damage. This review discusses the current literature reporting a role of lipids in αsyn misfolding and neurotoxicity in various synucleinopathy disorders and provides an overview of current methods to assess protein misfolding and pathogenicity both in vitro and in vivo.
Collapse
Affiliation(s)
- Cathryn L Ugalde
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia, .,the Departments of Microbiology and Immunology and.,the Howard Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia, and
| | | | - David I Finkelstein
- the Howard Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Andrew F Hill
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia, .,Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia, and
| |
Collapse
|
26
|
Khoshi A, Goodarzi G, Mohammadi R, Arezumand R, Moghbeli M, Najariyan M. Reducing effect of insulin resistance on alpha-synuclein gene expression in skeletal muscle. Diabetol Metab Syndr 2019; 11:99. [PMID: 31827624 PMCID: PMC6889442 DOI: 10.1186/s13098-019-0499-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/25/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Alpha-synuclein (SNCA) as the presynaptic protein is expressed in different tissues and prevents insulin-resistance (IR) through increasing glucose-uptake by adipocytes and muscles. However, the effect of insulin metabolism on SNCA expression has scarcely elucidated. In present study we assessed the probable effect of insulin resistance on SNCA expression in muscle C2C12 cells and also skeletal muscle tissues of type 2 diabetic mice. MATERIALS AND METHODS Sixteen male C57BL/6 mice were divided into two experimental groups, including control and type 2 diabetic mice with IR (induced by high-fat diet + low-dose streptozotocin). The animals of the study involved the measurements of fasting blood glucose, oral-glucose-tolerance-test, as well as fasting plasma insulin. Moreover, insulin-resistant and insulin-sensitive muscle C2C12 cells were prepared. The insulin-resistance was confirmed by the glucose-uptake assay. Comparative quantitative real time PCR was used to assess the SNCA expression. RESULTS The obtained results have showed a significant ~ 27% decrease in SNCA expression level in muscle tissue of diabetic mice (P = 0.022). Moreover, there was a significant change of SNCA expression in insulin-resistant C2C12 cells (P < 0.001). CONCLUSION Type 2 diabetes due to insulin-resistance can decrease SNCA gene expression in muscles. In addition to the role of SNCA in cell susceptibility to insulin and glucose uptake, the SNCA expression can also be affected by insulin metabolism.
Collapse
Affiliation(s)
- Amirhosein Khoshi
- Department of Clinical Biochemistry, School of Medicine, North Khorasan University of Medical Sciences, Arkan Roadway, Bojnurd, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of Medicine, North Khorasan University of Medical Sciences, Arkan Roadway, Bojnurd, Iran
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Mohammadi
- Department of Biotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Biotechnology, School of Medicine, Shaheed Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghaye Arezumand
- Department of Biotechnology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahnaz Najariyan
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
27
|
Requejo-Aguilar R, Bolaños JP. Mitochondrial control of cell bioenergetics in Parkinson's disease. Free Radic Biol Med 2016; 100:123-137. [PMID: 27091692 PMCID: PMC5065935 DOI: 10.1016/j.freeradbiomed.2016.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra. The earliest biochemical signs of the disease involve failure in mitochondrial-endoplasmic reticulum cross talk and lysosomal function, mitochondrial electron chain impairment, mitochondrial dynamics alterations, and calcium and iron homeostasis abnormalities. These changes are associated with increased mitochondrial reactive oxygen species (mROS) and energy deficiency. Recently, it has been reported that, as an attempt to compensate for the mitochondrial dysfunction, neurons invoke glycolysis as a low-efficient mode of energy production in models of PD. Here, we review how mitochondria orchestrate the maintenance of cellular energetic status in PD, with special focus on the switch from oxidative phosphorylation to glycolysis, as well as the implication of endoplasmic reticulum and lysosomes in the control of bioenergetics.
Collapse
Affiliation(s)
- Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Institute Maimonides of Biomedical Investigation of Cordoba (IMIBIC), Cordoba, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC, Zacarias Gonzalez, 2, 37007 Salamanca, Spain.
| |
Collapse
|
28
|
Guerreiro PS, Coelho JE, Sousa-Lima I, Macedo P, Lopes LV, Outeiro TF, Pais TF. Mutant A53T α-Synuclein Improves Rotarod Performance Before Motor Deficits and Affects Metabolic Pathways. Neuromolecular Med 2016; 19:113-121. [PMID: 27535567 DOI: 10.1007/s12017-016-8435-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
Abstract
The protein α-synuclein (α-Syn) interferes with glucose and lipid uptake and also activates innate immune cells. However, it remains unclear whether α-Syn or its familial mutant forms contribute to metabolic alterations and inflammation in synucleinopathies, such as Parkinson's disease (PD). Here, we address this issue in transgenic mice for the mutant A53T human α-Syn (α-SynA53T), a mouse model of synucleinopathies. At 9.5 months of age, mice overexpressing α-SynA53T (homozygous) had a significant reduction in weight, exhibited improved locomotion and did not show major motor deficits compared with control transgenic mice (heterozygous). At 17 months of age, α-SynA53T overexpression promoted general reduction in grip strength and deficient hindlimb reflex and resulted in severe disease and mortality in 50 % of the mice. Analysis of serum metabolites further revealed decreased levels of cholesterol, triglycerides and non-esterified fatty acids (NEFA) in α-SynA53T-overexpressing mice. In fed conditions, these mice also showed a significant decrease in serum insulin without alterations in blood glucose. In addition, assessment of inflammatory gene expression in the brain showed a significant increase in TNF-α mRNA but not of IL-1β induced by α-SynA53T overexpression. Interestingly, the brain mRNA levels of Sirtuin 2 (Sirt2), a deacetylase involved in both metabolic and inflammatory pathways, were significantly reduced. Our findings highlight the relevance of the mechanisms underlying initial weight loss and hyperactivity as early markers of synucleinopathies. Moreover, we found that changes in blood metabolites and decreased brain Sirt2 gene expression are associated with motor deficits.
Collapse
Affiliation(s)
- Patrícia S Guerreiro
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal.,VIB Switch Laboratory, Flanders Institute for Biotechnology (VIB), Leuven, 3000, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joana E Coelho
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal
| | - Inês Sousa-Lima
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal
| | - Paula Macedo
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.,APDP - Associação Protectora dos Diabéticos de Portugal, Rua Rodrigo da Fonseca, 1, 1250-189, Lisbon, Portugal
| | - Luísa V Lopes
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal
| | - Tiago F Outeiro
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.,Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Teresa F Pais
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisbon, Portugal. .,Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156, Oeiras, Portugal.
| |
Collapse
|
29
|
Emamzadeh FN. Alpha-synuclein structure, functions, and interactions. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2016; 21:29. [PMID: 27904575 PMCID: PMC5122110 DOI: 10.4103/1735-1995.181989] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 01/03/2016] [Accepted: 02/24/2016] [Indexed: 12/01/2022]
Abstract
At present, when a clinical diagnosis of Parkinson's disease (PD) is made, serious damage has already been done to nerve cells of the substantia nigra pars compacta. The diagnosis of PD in its earlier stages, before this irreversible damage, would be of enormous benefit for future treatment strategies designed to slow or halt the progression of this disease that possibly prevents accumulation of toxic aggregates. As a molecular biomarker for the detection of PD in its earlier stages, alpha-synuclein (α-syn), which is a key component of Lewy bodies, in which it is found in an aggregated and fibrillar form, has attracted considerable attention. Here, α-syn is reviewed in details.
Collapse
Affiliation(s)
- Fatemeh Nouri Emamzadeh
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, University of Lancaster, Lancaster, LA1 4AY, UK
| |
Collapse
|
30
|
Kuo T, Kim-Muller JY, McGraw TE, Accili D. Altered Plasma Profile of Antioxidant Proteins as an Early Correlate of Pancreatic β Cell Dysfunction. J Biol Chem 2016; 291:9648-56. [PMID: 26917725 DOI: 10.1074/jbc.m115.702183] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance and β cell dysfunction contribute to the pathogenesis of type 2 diabetes. Unlike insulin resistance, β cell dysfunction remains difficult to predict and monitor, because of the inaccessibility of the endocrine pancreas, the integrated relationship with insulin sensitivity, and the paracrine effects of incretins. The goal of our study was to survey the plasma response to a metabolic challenge in order to identify factors predictive of β cell dysfunction. To this end, we combined (i) the power of unbiased iTRAQ (isobaric tag for relative and absolute quantification) mass spectrometry with (ii) direct sampling of the portal vein following an intravenous glucose/arginine challenge (IVGATT) in (iii) mice with a genetic β cell defect. By so doing, we excluded the effects of peripheral insulin sensitivity as well as those of incretins on β cells, and focused on the first phase of insulin secretion to capture the early pathophysiology of β cell dysfunction. We compared plasma protein profiles with ex vivo islet secretome and transcriptome analyses. We detected changes to 418 plasma proteins in vivo, and detected changes to 262 proteins ex vivo The impairment of insulin secretion was associated with greater overall changes in the plasma response to IVGATT, possibly reflecting metabolic instability. Reduced levels of proteins regulating redox state and neuronal stress markers, as well as increased levels of coagulation factors, antedated the loss of insulin secretion in diabetic mice. These results suggest that a reduced complement of antioxidants in response to a mixed secretagogue challenge is an early correlate of future β cell failure.
Collapse
Affiliation(s)
- Taiyi Kuo
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Ja Young Kim-Muller
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Timothy E McGraw
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065
| | - Domenico Accili
- From the Department of Medicine and Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| |
Collapse
|
31
|
Bergström AL, Kallunki P, Fog K. Development of Passive Immunotherapies for Synucleinopathies. Mov Disord 2015; 31:203-13. [PMID: 26704735 DOI: 10.1002/mds.26481] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/07/2015] [Accepted: 10/15/2015] [Indexed: 01/13/2023] Open
Abstract
Immunotherapy using antibodies targeting alpha-synuclein has proven to be an effective strategy for ameliorating pathological and behavioral deficits induced by excess pathogenic alpha-synuclein in various animal and/or cellular models. However, the process of selecting the anti-alpha-synuclein antibody with the best potential to treat synucleinopathies in humans is not trivial. Critical to this process is a better understanding of the pathological processes involved in the synucleinopathies and how antibodies are able to influence these. We will give an overview of the first proof-of-concept studies in rodent disease models and discuss challenges associated with developing antibodies against alpha-synuclein resulting from the distribution and structural characteristics of the protein. We will also provide a status on the passive immunization approaches targeting alpha-synuclein that have entered, or are expected to enter, clinical evaluation.
Collapse
Affiliation(s)
| | - Pekka Kallunki
- Division of Neurodegeneration and Biologics, H. Lundbeck A/S, Valby, Denmark
| | - Karina Fog
- Division of Neurodegeneration and Biologics, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|