1
|
Ntetsika T, Catrina SB, Markaki I. Understanding the link between type 2 diabetes mellitus and Parkinson's disease: role of brain insulin resistance. Neural Regen Res 2025; 20:3113-3123. [PMID: 39715083 PMCID: PMC11881720 DOI: 10.4103/nrr.nrr-d-23-01910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/22/2024] [Accepted: 03/03/2024] [Indexed: 12/25/2024] Open
Abstract
Type 2 diabetes mellitus and Parkinson's disease are chronic diseases linked to a growing pandemic that affects older adults and causes significant socio-economic burden. Epidemiological data supporting a close relationship between these two aging-related diseases have resulted in the investigation of shared pathophysiological molecular mechanisms. Impaired insulin signaling in the brain has gained increasing attention during the last decade and has been suggested to contribute to the development of Parkinson's disease through the dysregulation of several pathological processes. The contribution of type 2 diabetes mellitus and insulin resistance in neurodegeneration in Parkinson's disease, with emphasis on brain insulin resistance, is extensively discussed in this article and new therapeutic strategies targeting this pathological link are presented and reviewed.
Collapse
Affiliation(s)
- Theodora Ntetsika
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Diabetes, Academic Specialist Center, Stockholm, Sweden
| | - Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| |
Collapse
|
2
|
Stockmann O, Ye L, Greten S, Chemodanow D, Wegner F, Klietz M. Impact of diabetes mellitus type two on incidence and progression of Parkinson's disease: a systematic review of longitudinal patient cohorts. J Neural Transm (Vienna) 2025; 132:627-635. [PMID: 39847072 PMCID: PMC12043777 DOI: 10.1007/s00702-025-02882-7] [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: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease of the elderly. Patients suffer from progressive motor and non-motor symptoms. Further, PD patients often present geriatric features like multimorbidity and polypharmacotherapy. A frequent comorbidity of PD patients is diabetes mellitus type two (T2DM). In the last decade growing evidence emerged on the impact of T2DM on PD. Of the present review was to analyze the impact of T2DM on PD incidence and progression in patient cohorts. A systematic review of the literature was performed via PubMed and Google Scholar. Studies on longitudinal PD patient cohorts with at least 10 patients per group were included. The diabetic state of the patient had to be determined. In total, 15 studies were analyzed for this review. According to most of the included studies T2DM increases the risk of developing PD significantly. Disease progression is augmented by T2DM both for motor and cognitive impairments. Some studies also point out a correlation of motor worsening and diabetic status measured by the serum HbA1c level. In relation to biomarkers, PD patients with diabetes have higher neurofilament light chain and Tau level but lower Amyloid beta level. T2DM seems to be a risk factor for the development and progression of PD. PD patients should be screened for T2DM and treatment should be initiated promptly. There is still a lack of knowledge about the molecular mechanisms leading to interactions of these diseases.
Collapse
Affiliation(s)
- Olga Stockmann
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Lan Ye
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Stephan Greten
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - David Chemodanow
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Martin Klietz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
3
|
Hornung S, Vogl DP, Naltsas D, Volta BD, Ballmann M, Marcon B, Syed MMK, Wu Y, Spanopoulou A, Feederle R, Heidrich L, Bernhagen J, Koeglsperger T, Höglinger GU, Rammes G, Lashuel HA, Kapurniotu A. Multi-Targeting Macrocyclic Peptides as Nanomolar Inhibitors of Self- and Cross-Seeded Amyloid Self-Assembly of α-Synuclein. Angew Chem Int Ed Engl 2025; 64:e202422834. [PMID: 39822034 DOI: 10.1002/anie.202422834] [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: 11/23/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Amyloid self-assembly of α-synuclein (αSyn) is linked to the pathogenesis of Parkinson's disease (PD). Type 2 diabetes (T2D) has recently emerged as a risk factor for PD. Cross-interactions between their amyloidogenic proteins may act as molecular links. In fact, fibrils of islet amyloid polypeptide (IAPP) (T2D) can cross-seed αSyn amyloidogenesis and αSyn and IAPP colocalize in PD brains. Inhibition of both self- and IAPP-cross-seeded αSyn amyloidogenesis could thus interfere with PD pathogenesis. Here we show that macrocyclic peptides, designed to mimic IAPP self-/cross-interaction sites and previously found to inhibit amyloidogenesis of IAPP and/or Alzheimer's disease (AD) amyloid-β peptide Aβ40(42), are nanomolar inhibitors of both self- and IAPP-cross-seeded amyloid self-assembly of αSyn. Anti-amyloid function is mediated by nanomolar affinity interactions with αSyn via three αSyn regions which are identified as key sites of both αSyn self-assembly and its cross-interactions with IAPP. We also show that the peptides block Aβ42-mediated cross-seeding of αSyn as well. Based on their broad spectrum anti-amyloid function and additional drug-like features, these peptides are leads for multifunctional anti-amyloid drugs in PD, T2D, AD, and their comorbidities, while the identified αSyn key segments are valuable targets for novel, multi-site targeting amyloid inhibitors in PD and related synucleinopathies.
Collapse
Affiliation(s)
- Simon Hornung
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Dominik P Vogl
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, Boehringer Ingelheim, Vienna, Austria
| | - Denise Naltsas
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Beatrice Dalla Volta
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Markus Ballmann
- Department of Anesthesiology and Intensive Care, Technische Universität München (TUM)/, Klinikum Rechts der Isar, Ismaningerstr. 22, D-81675, Munich, Germany
| | - Beatrice Marcon
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Muhammed Muazzam Kamil Syed
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Lausanne, Switzerland
| | - Yiyang Wu
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Anna Spanopoulou
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, ITM Isotope Technologies Munich SE, Garching/Munich, Germany
| | - Regina Feederle
- Core Facility Monoclonal Antibodies, Helmholtz Center Munich German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Luzia Heidrich
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, Life & Brain GmbH, Bonn, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilian-University (LMU), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Thomas Koeglsperger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care, Technische Universität München (TUM)/, Klinikum Rechts der Isar, Ismaningerstr. 22, D-81675, Munich, Germany
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Lausanne, Switzerland
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| |
Collapse
|
4
|
Waheed I, Sikandri T, Zaheen S, Khakwani MMAK, An Z, Liu T, Zhu C, Wei J. Evaluating the Molecular Interactions between Type 2 Diabetes Mellitus and Parkinson's Disease: Role of Antidiabetic Drugs as Promising Therapeutics. ACS Chem Neurosci 2025; 16:988-999. [PMID: 40042145 DOI: 10.1021/acschemneuro.4c00819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Evidence from previous research demonstrates a relationship between diabetes mellitus (DM) and Parkinson's disease (PD). T2DM is associated with chronic glucose dysregulation, as an etiological factor. It inhibits neuronal function through disrupted insulin signaling and oxidative stress, which ultimately lead to the loss of dopaminergic neurons in the substantia nigra (SN). Interactions between T2DM and PD were analyzed by gene expression, coexpression, and gene set enrichment via NCBI and STRING databases following pathways like KEGG and Reactome. The study identified nine key gene interactions through published literature on different databases and search engines that are involved in the progression of these chronic diseases. Furthermore, some genetic and nongenetic risk factors, gene mutations and environmental factors, are also involved in the progression of T2DM and PD. This review highlights the limitations of currently available drug treatments for these diseases and examines modern therapeutic approaches to address neurodegenerative and metabolic abnormalities. We critically assess the current experimental methodologies aimed at unraveling the pathophysiological mechanisms linking PD and T2DM while addressing the key challenges impeding a comprehensive understanding of the concurrent emergence of these debilitating age-related conditions.
Collapse
Affiliation(s)
- Irum Waheed
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Talal Sikandri
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Sumbal Zaheen
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | | | - Zhaowu An
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
5
|
Otzen DE, Peña-Díaz S, Widmann J, Daugberg AOH, Zhang Z, Jiang Y, Mittal C, Dueholm MKD, Louros N, Wang H, Javed I. Interactions between pathological and functional amyloid: A match made in Heaven or Hell? Mol Aspects Med 2025; 103:101351. [PMID: 40024004 DOI: 10.1016/j.mam.2025.101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
The amyloid state of proteins occurs in many different contexts in Nature and in modern society, ranging from the pathological kind (neurodegenerative diseases and amyloidosis) via man-made forms (food processing and - to a much smaller extent - protein biologics) to functional versions (bacterial biofilm, peptide hormones and signal transmission). These classes all come together in the human body which endogenously produces amyloidogenic protein able to form pathological human amyloid (PaHA), hosts a microbiome which continuously makes functional bacterial amyloid (FuBA) and ingests food which can contain amyloid. This can have grave consequences, given that PaHA can spread throughout the body in a "hand-me-down" fashion from cell to cell through small amyloid fragments, which can kick-start growth of new amyloid wherever they encounter monomeric amyloid precursors. Amyloid proteins can also self- and cross-seed across dissimilar peptide sequences. While it is very unlikely that ingested amyloid plays a role in this crosstalk, FuBA-PaHA interactions are increasingly implicated in vivo amyloid propagation. We are now in a position to understand the structural and bioinformatic basis for this cross-talk, thanks to the very recently obtained atomic-level structures of the two major FuBAs CsgA (E. coli) and FapC (Pseudomonas). While there are many reports of homology-driven heterotypic interactions between different PaHA, the human proteome does not harbor significant homology to CsgA and FapC. Yet we and others have uncovered significant cross-stimulation (and in some cases inhibition) of FuBA and PaHA both in vitro and in vivo, which we here rationalize based on structure and sequence. These interactions have important consequences for the transmission and development of neurodegenerative diseases, not least because FuBA and PaHA can come into contact via the gut-brain interface, recurrent infections with microbes and potentially even through invasive biofilm in the brain. Whether FuBA and PaHA first interact in the gut or the brain, they can both stimulate and block each other's aggregation as well as trigger inflammatory responses. The microbiome may also affect amyloidogenesis in other ways, e.g. through their own chaperones which recognize and block growth of both PaHA and FuBA as we show both experimentally and computationally. Heterotypic interactions between and within PaHA and FuBA both in vitro and in vivo are a vital part of the amyloid phenomenon and constitute a vibrant and exciting frontier for future research.
Collapse
Affiliation(s)
- Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| | - Samuel Peña-Díaz
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| | - Jeremias Widmann
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Anders Ogechi Hostrup Daugberg
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg OE, Denmark
| | - Zhefei Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yanting Jiang
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Chandrika Mittal
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Morten K D Dueholm
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg OE, Denmark
| | - Nikolaos Louros
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA; Department of Biophysics, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Huabing Wang
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China; Jiangsu Fuyuda Food Products Co., Ltd, Qinyou Road 88, Gaoyou City, Jiangsu Province, 225600, China.
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld, 4072, Australia.
| |
Collapse
|
6
|
Wang Y, Gao P, Wu Z, Jiang B, Wang Y, He Z, Zhao B, Tian X, Gao H, Cai L, Li W. Exploring the therapeutic potential of Chinese herbs on comorbid type 2 diabetes mellitus and Parkinson's disease: A mechanistic study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119095. [PMID: 39537117 DOI: 10.1016/j.jep.2024.119095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) and Parkinson's disease (PD) are chronic conditions that affect the aging population, with increasing prevalence globally. The rising prevalence of comorbidity between these conditions, driven by demographic shifts, severely impacts the quality of life of patients, posing a significant burden on healthcare resources. Chinese herbal medicine has been used to treat T2DM and PD for millennia. Pharmacological studies have demonstrated that medicinal herbs effectively lower blood glucose levels and exert neuroprotective effects, suggesting their potential as adjunctive therapy for concurrent management of T2DM and PD. AIM OF THE STUDY To elucidate the shared mechanisms underlying T2DM and PD, particularly focusing on the potential mechanisms by which medicinal herbs (including herbal formulas, single herbs, and active compounds) may treat these diseases, to provide valuable insights for developing therapeutics targeting comorbid T2DM and PD. MATERIALS AND METHODS Studies exploring the mechanisms underlying T2DM and PD, as well as the treatment of these conditions with medicinal herbs, were extracted from several electronic databases, including PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure (CNKI). RESULTS Numerous studies have shown that inflammation, oxidative stress, insulin resistance, impaired autophagy, gut microbiota dysbiosis, and ferroptosis are shared mechanisms underlying T2DM and PD mediated through the NLRP3 inflammasome, NF-κB, MAPK, Keap1/Nrf2/ARE, PI3K/AKT, AMPK/SIRT1, and System XC--GSH-GPX4 signaling pathways. Thirty-four medicinal herbs, including 2 herbal formulas, 4 single herbs, and 28 active compounds, have been reported to potentially exert anti-T2DM and anti-PD effects by targeting these shared mechanisms. CONCLUSIONS Traditional Chinese medicine effectively combats T2DM and PD through shared pathological mechanisms, highlighting their potential for application in treating these comorbid conditions.
Collapse
Affiliation(s)
- Yan Wang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Pengpeng Gao
- Department of Preventive Treatment, Ningxia Integrated Chinese and Western Medicine Hospital, Yinchuan, 750004, China
| | - Zicong Wu
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhaxicao He
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bing Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinyun Tian
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Han Gao
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Cai
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
7
|
Tur EK, Kutlu HB, Sever N, Arı BC, Gözke E. Increased dementia risk in patients with Parkinson's disease attributed to metabolic syndrome. Neurol Sci 2025; 46:733-741. [PMID: 39470904 DOI: 10.1007/s10072-024-07803-2] [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: 08/16/2024] [Accepted: 10/02/2024] [Indexed: 11/01/2024]
Abstract
INTRODUCTION Metabolic syndrome (MetS) manifests resembling pathophysiological mechanisms with Parkinson's disease (PD). Current research on the overall population has emphasized MetS as a freestanding risk factor for cognition. This research aims to explore the impact of MetS on cognition in Parkinson's patients. METHOD We involved subjects identified as having early-stage PD patients. The MetS was diagnosed dependent on parameters overviewed in the National Cholesterol Education Program's Adult Treatment Panel III. The clinical severity and stages in patients with PD were dependent on the disease rating scales. The cognition was evaluated by the Turkısh version of the Montreal Cognitive Assessment Scale (MoCA-TR). The cases were categorized according to cognitive failure: mild cognitive impairment in PD (PD-MCI), and PD dementia (PDD). RESULTS Metabolic syndrome was present in 39.6% of the participants. 22.0% of patients were in the normal cognition, 29.1% in the PD-MCI group, and 48.9% in the PD-D group. The cognitive scores in patients with MetS is considerably lower than MetS negative group. A statistically notable inverse association was detected between fasting blood glucose levels and the visual-spatial/executive functions, naming, language, and orientation scores. The multivariate logistic regression analysis showed individuals with MetS were found to have an 11.308 times higher risk of PD-D (odds ratio [OR]: 11,3, 95% confidence interval [CI]: 1.61-79.2 ). CONCLUSION We discerned the occurrence of MetS in PD raises the possibility of advancing dementia. This suggests that considering MetS in this patient group could contribute to the effective management of dementia.
Collapse
Affiliation(s)
- Esma Kobak Tur
- Department of Neurology, University of Health Sciences, Fatih Sultan Mehmet Research and Training Hospital, Hastane Sok. No: 1/8 Içerenköy - Ataşehir, Istanbul, 34752, Turkey.
| | - Helin Berfin Kutlu
- Department of Neurology, University of Health Sciences, Fatih Sultan Mehmet Research and Training Hospital, Hastane Sok. No: 1/8 Içerenköy - Ataşehir, Istanbul, 34752, Turkey
| | - Nisa Sever
- Department of Neurology, University of Health Sciences, Fatih Sultan Mehmet Research and Training Hospital, Hastane Sok. No: 1/8 Içerenköy - Ataşehir, Istanbul, 34752, Turkey
| | - Buse Cagla Arı
- Department of Neurology, Bahcesehir University Medical Faculty, Istanbul, Turkey
| | - Eren Gözke
- Department of Neurology, University of Health Sciences, Fatih Sultan Mehmet Research and Training Hospital, Hastane Sok. No: 1/8 Içerenköy - Ataşehir, Istanbul, 34752, Turkey
| |
Collapse
|
8
|
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
|
9
|
Zhang RY, Li JY, Liu YN, Zhang ZX, Zhao J, Li FJ. The causal relationship between type 2 diabetes mellitus and isolated REM sleep behavior disorder: results from multivariable and network Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1408053. [PMID: 39655344 PMCID: PMC11625559 DOI: 10.3389/fendo.2024.1408053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/25/2024] [Indexed: 12/12/2024] Open
Abstract
Objectives To investigate the causal relationship between type 2 diabetes mellitus (T2DM, exposure) and isolated REM sleep behavior disorder (iRBD, outcome). Methods Genome-wide association study (GWAS) data for iRBD comprised 9,447 samples, including 1,061 iRBD cases from the International RBD Study Group. Initially, we performed linkage disequilibrium score regression (LDSC) to explore the genetic correlation between T2DM and iRBD. Then the two-sample univariate MR (UVMR) analysis was conducted to examine the effects of T2DM and blood sugar metabolism-related factors on iRBD. Subsequently, we applied multivariable MR (MVMR) methods to further adjust for confounders. Lastly, we executed a network MR analysis, with cytokines and immune cell characteristics as potential mediators, aiming to investigate indirect effect of T2DM on iRBD. Results Results from LDSC suggest a genetic correlation between T2DM and iRBD (rg=0.306, P=0.029). UVMR analysis indicates that both T2DM (Odds Ratio [95% Confidence Interval] = 1.19 [1.03, 1.37], P = 0.017) and high blood glucose levels (1.55 [1.04, 2.30], P = 0.032) are risk factors for iRBD. Even after adjusting for confounders in MVMR, the association between T2DM and iRBD remains robust. Finally, results from network MR analysis suggest that T2DM may indirectly promote the development of iRBD by reducing levels of Stromal Cell-Derived Factor 2 in circulation and by increasing BAFF-receptor expression in IgD- CD38- B cells. Conclusions T2DM may promote the onset of iRBD by influencing immune-inflammatory responses. Our findings provide valuable insights and directions for understanding the pathogenesis of iRBD, identifying high-risk groups, and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Ru-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, First People's Hospital of Zigong, Zigong, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Ning Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zi-Xuan Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fu-Jia Li
- Department of Pulmonary and Critical Care Medicine, First People's Hospital of Zigong, Zigong, Sichuan, China
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Ben Shaul T, Frenkel D, Gurevich T. The Interplay of Stress, Inflammation, and Metabolic Factors in the Course of Parkinson's Disease. Int J Mol Sci 2024; 25:12409. [PMID: 39596474 PMCID: PMC11594997 DOI: 10.3390/ijms252212409] [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: 09/29/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative condition for which there are symptomatic treatments but no disease-modifying therapies (DMTs). Extensive research over the years has highlighted the need for a multi-target DMT approach in PD that recognizes the various risk factors and their intricate interplay in contributing to PD-related neurodegeneration. Widespread risk factors, such as emotional stress and metabolic factors, have increasingly become focal points of exploration. Our review aims to summarize interactions between emotional stress and selected key players in metabolism, such as insulin, as potential mechanisms underlying neurodegeneration in PD.
Collapse
Affiliation(s)
- Tal Ben Shaul
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tanya Gurevich
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
11
|
Song Z, Tang H, Gatch A, Sun Y, Ding F. Islet amyloid polypeptide fibril catalyzes amyloid-β aggregation by promoting fibril nucleation rather than direct axial growth. Int J Biol Macromol 2024; 279:135137. [PMID: 39208885 PMCID: PMC11469950 DOI: 10.1016/j.ijbiomac.2024.135137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aberrant aggregation of amyloid-β (Aβ) and islet amyloid polypeptide (IAPP) into amyloid fibrils underlies the pathogenesis of Alzheimer's disease (AD) and type 2 diabetes (T2D), respectively. T2D significantly increases AD risk, with evidence suggesting that IAPP and Aβ co-aggregation and cross-seeding might contribute to the cross-talk between two diseases. Experimentally, preformed IAPP fibril seeds can accelerate Aβ aggregation, though the cross-seeding mechanism remains elusive. Here, we computationally demonstrated that Aβ monomer preferred to bind to the elongation ends of preformed IAPP fibrils. However, due to sequence mismatch, the Aβ monomer could not directly grow onto IAPP fibrils by forming multiple stable β-sheets with the exposed IAPP peptides. Conversely, in our control simulations of self-seeding, the Aβ monomer could axially grow on the Aβ fibril, forming parallel in-register β-sheets. Additionally, we showed that the IAPP fibril could catalyze Aβ fibril nucleation by promoting the formation of parallel in-register β-sheets in the C-terminus between bound Aβ peptides. This study enhances our understanding of the molecular interplay between Aβ and IAPP, shedding light on the cross-seeding mechanisms potentially linking T2D and AD. Our findings also underscore the importance of clearing IAPP deposits in T2D patients to mitigate AD risk.
Collapse
Affiliation(s)
- Zhiyuan Song
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States; Department of Engineering Mechanics, Hohai University, Nanjing 210098, China
| | - Adam Gatch
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States; School of Physical Science and Technology, Ningbo University, Ningbo 315211, China
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States.
| |
Collapse
|
12
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
13
|
Tang Y, Zhang Y, Zhang D, Liu Y, Nussinov R, Zheng J. Exploring pathological link between antimicrobial and amyloid peptides. Chem Soc Rev 2024; 53:8713-8763. [PMID: 39041297 DOI: 10.1039/d3cs00878a] [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: 07/24/2024]
Abstract
Amyloid peptides (AMYs) and antimicrobial peptides (AMPs) are considered as the two distinct families of peptides, characterized by their unique sequences, structures, biological functions, and specific pathological targets. However, accumulating evidence has revealed intriguing pathological connections between these peptide families in the context of microbial infection and neurodegenerative diseases. Some AMYs and AMPs share certain structural and functional characteristics, including the ability to self-assemble, the presence of β-sheet-rich structures, and membrane-disrupting mechanisms. These shared features enable AMYs to possess antimicrobial activity and AMPs to acquire amyloidogenic properties. Despite limited studies on AMYs-AMPs systems, the cross-seeding phenomenon between AMYs and AMPs has emerged as a crucial factor in the bidirectional communication between the pathogenesis of neurodegenerative diseases and host defense against microbial infections. In this review, we examine recent developments in the potential interplay between AMYs and AMPs, as well as their pathological implications for both infectious and neurodegenerative diseases. By discussing the current progress and challenges in this emerging field, this account aims to inspire further research and investments to enhance our understanding of the intricate molecular crosstalk between AMYs and AMPs. This knowledge holds great promise for the development of innovative therapies to combat both microbial infections and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yijing Tang
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Ohio 44325, USA.
| | - Yanxian Zhang
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Dong Zhang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
- Department of Human Molecular Genetics and Biochemistry Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jie Zheng
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Ohio 44325, USA.
| |
Collapse
|
14
|
Flint Z, Grannemann H, Baffour K, Koti N, Taylor E, Grier E, Sutton C, Johnson D, Dandawate P, Patel R, Santra S, Banerjee T. Mechanistic Insights Behind the Self-Assembly of Human Insulin under the Influence of Surface-Engineered Gold Nanoparticles. ACS Chem Neurosci 2024; 15:2359-2371. [PMID: 38728258 PMCID: PMC11157486 DOI: 10.1021/acschemneuro.4c00226] [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: 04/13/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Elucidating the underlying principles of amyloid protein self-assembly at nanobio interfaces is extremely challenging due to the diversity in physicochemical properties of nanomaterials and their physical interactions with biological systems. It is, therefore, important to develop nanoscale materials with dynamic features and heterogeneities. In this work, through engineering of hierarchical polyethylene glycol (PEG) structures on gold nanoparticle (GNP) surfaces, tailored nanomaterials with different surface properties and conformations (GNPs-PEG) are created for modulating the self-assembly of a widely studied protein, insulin, under amyloidogenic conditions. Important biophysical studies including thioflavin T (ThT) binding, circular dichroism (CD), surface plasmon resonance (SPR), and atomic force microscopy (AFM) showed that higher-molecular weight GNPs-PEG triggered the formation of amyloid fibrils by promoting adsorption of proteins at nanoparticle surfaces and favoring primary nucleation rate. Moreover, the modulation of fibrillation kinetics reduces the overall toxicity of insulin oligomers and fibrils. In addition, the interaction between the PEG polymer and amyloidogenic insulin examined using MD simulations revealed major changes in the secondary structural elements of the B chain of insulin. The experimental findings provide molecular-level descriptions of how the PEGylated nanoparticle surface modulates protein adsorption and drives the self-assembly of insulin. This facile approach provides a new avenue for systematically altering the binding affinities on nanoscale surfaces by tailoring their topologies for examining adsorption-induced fibrillogenesis phenomena of amyloid proteins. Together, this study suggests the role of nanobio interfaces during surface-induced heterogeneous nucleation as a primary target for designing therapeutic interventions for amyloid-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Zachary Flint
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Haylee Grannemann
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Kristos Baffour
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Neelima Koti
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Emma Taylor
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Ethan Grier
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Carissa Sutton
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - David Johnson
- Molecular
Graphics and Modeling Laboratory, University
of Kansas, 2034 Becker
Drive, Lawrence, Kansas 66018, United States
| | - Prasad Dandawate
- Department
of Cancer Biology, The University of Kansas
Medical Center, Kansas City, Kansas 66160, United States
| | - Rishi Patel
- Jordan
Valley Innovation Center, Missouri State
University, 542 N. Boonville
Avenue, Springfield, Missouri 65806, United States
| | - Santimukul Santra
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| | - Tuhina Banerjee
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States
| |
Collapse
|
15
|
Ye L, Greten S, Wegner F, Doll-Lee J, Krey L, Heine J, Gandor F, Vogel A, Berger L, Gruber D, Levin J, Katzdobler S, Peters O, Dashti E, Priller J, Spruth EJ, Kühn AA, Krause P, Spottke A, Schneider A, Beyle A, Kimmich O, Donix M, Haussmann R, Brandt M, Dinter E, Wiltfang J, Schott BH, Zerr I, Bähr M, Buerger K, Janowitz D, Perneczky R, Rauchmann BS, Weidinger E, Düzel E, Glanz W, Teipel S, Kilimann I, Wurster I, Brockmann K, Hoffmann DC, Klockgether T, Krause O, Heck J, Höglinger GU, Klietz M. The comorbidity profiles and medication issues of patients with multiple system atrophy: a systematic cross-sectional analysis. J Neurol 2024; 271:2639-2648. [PMID: 38353748 PMCID: PMC11055732 DOI: 10.1007/s00415-024-12207-5] [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: 12/01/2023] [Accepted: 01/16/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Multiple system atrophy (MSA) is a complex and fatal neurodegenerative movement disorder. Understanding the comorbidities and drug therapy is crucial for MSA patients' safety and management. OBJECTIVES To investigate the pattern of comorbidities and aspects of drug therapy in MSA patients. METHODS Cross-sectional data of MSA patients according to Gilman et al. (2008) diagnostic criteria and control patients without neurodegenerative diseases (non-ND) were collected from German, multicenter cohorts. The prevalence of comorbidities according to WHO ICD-10 classification and drugs administered according to WHO ATC system were analyzed. Potential drug-drug interactions were identified using AiDKlinik®. RESULTS The analysis included 254 MSA and 363 age- and sex-matched non-ND control patients. MSA patients exhibited a significantly higher burden of comorbidities, in particular diseases of the genitourinary system. Also, more medications were prescribed MSA patients, resulting in a higher prevalence of polypharmacy. Importantly, the risk of potential drug-drug interactions, including severe interactions and contraindicated combinations, was elevated in MSA patients. When comparing MSA-P and MSA-C subtypes, MSA-P patients suffered more frequently from diseases of the genitourinary system and diseases of the musculoskeletal system and connective tissue. CONCLUSIONS MSA patients face a substantial burden of comorbidities, notably in the genitourinary system. This, coupled with increased polypharmacy and potential drug interactions, highlights the complexity of managing MSA patients. Clinicians should carefully consider these factors when devising treatment strategies for MSA patients.
Collapse
Affiliation(s)
- Lan Ye
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stephan Greten
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Johanna Doll-Lee
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Lea Krey
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Johanne Heine
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Florin Gandor
- Neurologisches Fachkrankenhaus für Bewegungsstörungen/Parkinson, Kliniken Beelitz, 14547, Beelitz-Heilstätten, Germany
| | - Annemarie Vogel
- Neurologisches Fachkrankenhaus für Bewegungsstörungen/Parkinson, Kliniken Beelitz, 14547, Beelitz-Heilstätten, Germany
| | - Luise Berger
- Neurologisches Fachkrankenhaus für Bewegungsstörungen/Parkinson, Kliniken Beelitz, 14547, Beelitz-Heilstätten, Germany
| | - Doreen Gruber
- Neurologisches Fachkrankenhaus für Bewegungsstörungen/Parkinson, Kliniken Beelitz, 14547, Beelitz-Heilstätten, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Eman Dashti
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea A Kühn
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité, University Medicine Berlin, Charité, Berlin, Germany
| | - Patricia Krause
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité, University Medicine Berlin, Charité, Berlin, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Aline Beyle
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Markus Donix
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Robert Haussmann
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Moritz Brandt
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Dinter
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Göttingen, Germany
- Neurosciences and Signaling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Göttingen, Germany
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| | - Mathias Bähr
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University, Magdeburg, Germany
- Clinic for Neurology, Medical Faculty, University Hospital Magdeburg, Magdeburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Isabel Wurster
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Olaf Krause
- DIAKOVERE Henriettenstift and Department of General Medicine and Palliative Care, Center for Medicine of the Elderly, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Center for Geriatric Medicine, Hospital DIAKOVERE Henriettenstift, Schwe-Mannstrasse 19, 30559, Hannover, Germany
| | - Johannes Heck
- Institute for Clinical Pharmacology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Günter U Höglinger
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Martin Klietz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
16
|
Ribarič S. The Contribution of Type 2 Diabetes to Parkinson's Disease Aetiology. Int J Mol Sci 2024; 25:4358. [PMID: 38673943 PMCID: PMC11050090 DOI: 10.3390/ijms25084358] [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: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are chronic disorders that have a significant health impact on a global scale. Epidemiological, preclinical, and clinical research underpins the assumption that insulin resistance and chronic inflammation contribute to the overlapping aetiologies of T2D and PD. This narrative review summarises the recent evidence on the contribution of T2D to the initiation and progression of PD brain pathology. It also briefly discusses the rationale and potential of alternative pharmacological interventions for PD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
17
|
Li X, Zhang Y, Yang Z, Zhang S, Zhang L. The Inhibition Effect of Epigallocatechin-3-Gallate on the Co-Aggregation of Amyloid-β and Human Islet Amyloid Polypeptide Revealed by Replica Exchange Molecular Dynamics Simulations. Int J Mol Sci 2024; 25:1636. [PMID: 38338914 PMCID: PMC10855639 DOI: 10.3390/ijms25031636] [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: 01/04/2024] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's disease and Type 2 diabetes are two epidemiologically linked diseases which are closely associated with the misfolding and aggregation of amyloid proteins amyloid-β (Aβ) and human islet amyloid polypeptide (hIAPP), respectively. The co-aggregation of the two amyloid proteins is regarded as the fundamental molecular mechanism underlying their pathological association. The green tea extract epigallocatechin-3-gallate (EGCG) has been extensively demonstrated to inhibit the amyloid aggregation of Aβ and hIAPP proteins. However, its potential role in amyloid co-aggregation has not been thoroughly investigated. In this study, we employed the enhanced-sampling replica exchange molecular dynamics simulation (REMD) method to investigate the effect of EGCG on the co-aggregation of Aβ and hIAPP. We found that EGCG molecules substantially diminish the β-sheet structures within the amyloid core regions of Aβ and hIAPP in their co-aggregates. Through hydrogen-bond, π-π and cation-π interactions targeting polar and aromatic residues of Aβ and hIAPP, EGCG effectively attenuates both inter-chain and intra-chain interactions within the co-aggregates. All these findings indicated that EGCG can effectively inhibit the co-aggregation of Aβ and hIAPP. Our study expands the potential applications of EGCG as an anti-amyloidosis agent and provides therapeutic options for the pathological association of amyloid misfolding disorders.
Collapse
Affiliation(s)
- Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China (Z.Y.); (S.Z.); (L.Z.)
- State Key Laboratory of Surface Physics, Department of Physics, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Yu Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China (Z.Y.); (S.Z.); (L.Z.)
| | - Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China (Z.Y.); (S.Z.); (L.Z.)
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China (Z.Y.); (S.Z.); (L.Z.)
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China (Z.Y.); (S.Z.); (L.Z.)
| |
Collapse
|
18
|
Li Z, Jiang X, Yang M, Pan Y. Association between falls and nonmotor symptoms in patients with Parkinson's disease. J Clin Neurosci 2023; 118:143-146. [PMID: 37939511 DOI: 10.1016/j.jocn.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 09/24/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Parkinson's disease (PD) is a chronic neurodegenerative disorder. Falls are common in patients with PD and can lead to disability, bedridden status, and death. The mechanisms of falls induced by symptoms of PD have not been fully clarified. We investigated the association between falls and nonmotor symptoms in PD patients. METHODS A total of 361 patients with Parkinson's disease were included. Whether the patients had fallen in the past half a year was recorded. Nonmotor symptoms were assessed by 30 items from the nonmotor symptom questionnaire (NMS Quest), Parkinson's Disease Sleep Scale (PDSS), Hamilton Depression Scale (HAMD), Hamilton Anxiety Scale (HAMA), and Montreal Cognitive Assessment Scale (MOCA). RESULTS A total of 63 patients experienced falls in the past six months, with an incidence of 17.5%. The patients with falls were elderly, had severe motor symptoms and disease severity, and the proportion of diabetic patients who experienced falls was higher. Adjusted for the above factors, the results showed that patients with falls had higher PD-NMS, HAMD and HAMA scores, but there was no significant difference in the total score and subscores of the MoCA scale between the two groups. The risk factors related to falling included age, history of diabetes, depression (HAMD), HAMD cognitive impairment, NMS urinary tract and NMS postural hypotension. CONCLUSIONS Falls were a common symptom in patients with PD and were not only related to motor symptoms but also closely related to nonmotor symptoms. urinary tract symptoms, postural hypotension, depression and HAMD cognitive impairment were risk factors related to falling in patients with PD.
Collapse
Affiliation(s)
- Zhen Li
- Department of Geriatric Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xu Jiang
- Department of Geriatric Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Minggang Yang
- Department of Neurology, Xuyi People's Hospital, Xuyi, Jiangsu 211700, China
| | - Yang Pan
- Department of Geriatric Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
19
|
Vijiaratnam N, Foltynie T. How should we be using biomarkers in trials of disease modification in Parkinson's disease? Brain 2023; 146:4845-4869. [PMID: 37536279 PMCID: PMC10690028 DOI: 10.1093/brain/awad265] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson's disease has formed the backbone for a proposed staging system for incorporation in Parkinson's disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson's disease patients into trials (as distinct from patients with non-Parkinson's disease parkinsonism or non-Parkinson's disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson's disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer's disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson's disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson's disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson's disease.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
20
|
Rani K, Pal A, Gurnani B, Agarwala P, Sasmal DK, Jain N. An Innate Host Defense Protein β 2-Microglobulin Keeps a Check on α-Synuclein amyloid Assembly: Implications in Parkinson's Disease. J Mol Biol 2023; 435:168285. [PMID: 37741548 DOI: 10.1016/j.jmb.2023.168285] [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: 05/10/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
Amyloid formation due to protein misfolding has gained significant attention due to its association with neurodegenerative diseases. α-Synuclein (α-syn) is one such protein that undergoes a profound conformational switch to form higher order cross-β-sheet structures, resulting in amyloid formation, which is linked to the pathophysiology of Parkinson's disease (PD). The present status of research on α-syn aggregation and PD reveals that the disease progression may be linked with many other diseases, such as kidney-related disorders. Unraveling the link between PD and non-neurological diseases may help in early detection and a better understanding of PD progression. Herein, we investigated the modulation of α-syn in the presence of β2-microglobulin (β2m), a structural protein associated with dialysis-related amyloidosis. We took a multi-disciplinary approach to establish that β2m mitigates amyloid formation by α-syn. Our fluorescence, microscopy and toxicity data demonstrated that sub-stoichiometric ratio of β2m drives α-syn into off-pathway non-toxic aggregates incompetent of transforming into amyloids. Using AlphaFold2 and all-atom MD simulation, we showed that the β-strand segments (β1 and β2) of α-synuclein, which frequently engage in interactions within amyloid fibrils, interact with the last β-strand at the C-terminal of β2m. The outcome of this study will unravel the yet unknown potential linkage of PD with kidney-related disorders. Insights from the cross-talk between two amyloidogenic proteins will lead to early diagnosis and new therapeutic approaches for treating Parkinson's disease. Finally, disruption of the nucleation process of α-syn amyloids by targeting the β1-β2 region will constitute a potential therapeutic approach for inhibiting amyloid formation.
Collapse
Affiliation(s)
- Khushboo Rani
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Rajasthan, India. https://twitter.com/khushboo251995
| | - Arumay Pal
- School of Bioengineering, Vellore Institute of Technology, Bhopal, India. https://twitter.com/Arumay_Pal
| | - Bharat Gurnani
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Rajasthan, India. https://twitter.com/bgurnani05
| | - Pratibha Agarwala
- Department of Chemistry, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Rajasthan, India
| | - Dibyendu K Sasmal
- Department of Chemistry, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Rajasthan, India
| | - Neha Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Rajasthan, India; Centre for Emerging Technologies for Sustainable Development (CETSD), Indian Institute of Technology Jodhpur, Nagaur Road, Karwar 342030, Rajasthan, India.
| |
Collapse
|
21
|
Heid LF, Kupreichyk T, Schützmann MP, Schneider W, Stoldt M, Hoyer W. Nucleation of α-Synuclein Amyloid Fibrils Induced by Cross-Interaction with β-Hairpin Peptides Derived from Immunoglobulin Light Chains. Int J Mol Sci 2023; 24:16132. [PMID: 38003322 PMCID: PMC10671648 DOI: 10.3390/ijms242216132] [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: 09/15/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Heterologous interactions between different amyloid-forming proteins, also called cross-interactions, may have a critical impact on disease-related amyloid formation. β-hairpin conformers of amyloid-forming proteins have been shown to affect homologous interactions in the amyloid self-assembly process. Here, we applied two β-hairpin-forming peptides derived from immunoglobulin light chains as models to test how heterologous β-hairpins modulate the fibril formation of Parkinson's disease-associated protein α-synuclein (αSyn). The peptides SMAhp and LENhp comprise β-strands C and C' of the κ4 antibodies SMA and LEN, which are associated with light chain amyloidosis and multiple myeloma, respectively. SMAhp and LENhp bind with high affinity to the β-hairpin-binding protein β-wrapin AS10 according to isothermal titration calorimetry and NMR spectroscopy. The addition of SMAhp and LENhp affects the kinetics of αSyn aggregation monitored by Thioflavin T (ThT) fluorescence, with the effect depending on assay conditions, salt concentration, and the applied β-hairpin peptide. In the absence of agitation, substoichiometric concentrations of the hairpin peptides strongly reduce the lag time of αSyn aggregation, suggesting that they support the nucleation of αSyn amyloid fibrils. The effect is also observed for the aggregation of αSyn fragments lacking the N-terminus or the C-terminus, indicating that the promotion of nucleation involves the interaction of hairpin peptides with the hydrophobic non-amyloid-β component (NAC) region.
Collapse
Affiliation(s)
- Laetitia F. Heid
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Tatsiana Kupreichyk
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Marie P. Schützmann
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Walfried Schneider
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Matthias Stoldt
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
- Institute of Biological Information Processing (IBI-7) and JuStruct, Jülich Center for Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
22
|
Meng L, Liu C, Li Y, Chen G, Xiong M, Yu T, Pan L, Zhang X, Zhou L, Guo T, Yuan X, Liu C, Zhang Z, Zhang Z. The yeast prion protein Sup35 initiates α-synuclein pathology in mouse models of Parkinson's disease. SCIENCE ADVANCES 2023; 9:eadj1092. [PMID: 37910610 PMCID: PMC10619926 DOI: 10.1126/sciadv.adj1092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023]
Abstract
Parkinson's disease (PD) is characterized by the pathologic aggregation and prion-like propagation of α-synuclein (α-syn). Emerging evidence shows that fungal infections increase the incidence of PD. However, the molecular mechanisms by which fungi promote the onset of PD are poorly understood. Here, we show that nasal infection with Saccharomyces cerevisiae (S. cerevisiae) in α-syn A53T transgenic mice accelerates the aggregation of α-syn. Furthermore, we found that Sup35, a prion protein from S. cerevisiae, is the key factor initiating α-syn pathology induced by S. cerevisiae. Sup35 interacts with α-syn and accelerates its aggregation in vitro. Notably, injection of Sup35 fibrils into the striatum of wild-type mice led to α-syn pathology and PD-like motor impairment. The Sup35-seeded α-syn fibrils showed enhanced seeding activity and neurotoxicity compared with pure α-syn fibrils in vitro and in vivo. Together, these observations indicate that the yeast prion protein Sup35 initiates α-syn pathology in PD.
Collapse
Affiliation(s)
- Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Congcong Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yiming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guiqin Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingyan Zhou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tao Guo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xin Yuan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chaoyang Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan 430073, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
23
|
Li LY, Liu SF, Zhuang JL, Li MM, Huang ZP, Chen YH, Chen XR, Chen CN, Lin S, Ye LC. Recent research progress on metabolic syndrome and risk of Parkinson's disease. Rev Neurosci 2023; 34:719-735. [PMID: 36450297 DOI: 10.1515/revneuro-2022-0093] [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: 07/28/2022] [Accepted: 11/06/2022] [Indexed: 10/05/2023]
Abstract
Parkinson's disease (PD) is one of the most widespread neurodegenerative diseases. PD is associated with progressive loss of substantia nigra dopaminergic neurons, including various motor symptoms (e.g., bradykinesia, rigidity, and resting tremor), as well as non-motor symptoms (e.g., cognitive impairment, constipation, fatigue, sleep disturbance, and depression). PD involves multiple biological processes, including mitochondrial or lysosomal dysfunction, oxidative stress, insulin resistance, and neuroinflammation. Metabolic syndrome (MetS), a collection of numerous connected cerebral cardiovascular conditions, is a common and growing public health problem associated with many chronic diseases worldwide. MetS components include central/abdominal obesity, systemic hypertension, diabetes, and atherogenic dyslipidemia. MetS and PD share multiple pathophysiological processes, including insulin resistance, oxidative stress, and chronic inflammation. In recent years, MetS has been linked to an increased risk of PD, according to studies; however, the specific mechanism remains unclear. Researchers also found that some related metabolic therapies are potential therapeutic strategies to prevent and improve PD. This article reviews the epidemiological relationship between components of MetS and the risk of PD and discusses the potentially relevant mechanisms and recent progress of MetS as a risk factor for PD. Furthermore, we conclude that MetS-related therapies are beneficial for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Lin-Yi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Shu-Fen Liu
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Jian-Long Zhuang
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou 362000, China
| | - Mi-Mi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Zheng-Ping Huang
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Yan-Hong Chen
- Department of Neurology, Shishi General Hospital, Quanzhou 362000, Fujian Province, China
| | - Xiang-Rong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Chun-Nuan Chen
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW, Australia
| | - Li-Chao Ye
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
| |
Collapse
|
24
|
Kakraba S, Ayyadevara S, Mainali N, Balasubramaniam M, Bowroju S, Penthala NR, Atluri R, Barger SW, Griffin ST, Crooks PA, Shmookler Reis RJ. Thiadiazolidinone (TDZD) Analogs Inhibit Aggregation-Mediated Pathology in Diverse Neurodegeneration Models, and Extend C. elegans Life- and Healthspan. Pharmaceuticals (Basel) 2023; 16:1498. [PMID: 37895969 PMCID: PMC10610358 DOI: 10.3390/ph16101498] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic, low-grade inflammation has been implicated in aging and age-dependent conditions, including Alzheimer's disease, cardiomyopathy, and cancer. One of the age-associated processes underlying chronic inflammation is protein aggregation, which is implicated in neuroinflammation and a broad spectrum of neurodegenerative diseases such as Alzheimer's, Huntington's, and Parkinson's diseases. We screened a panel of bioactive thiadiazolidinones (TDZDs) from our in-house library for rescue of protein aggregation in human-cell and C. elegans models of neurodegeneration. Among the tested TDZD analogs, PNR886 and PNR962 were most effective, significantly reducing both the number and intensity of Alzheimer-like tau and amyloid aggregates in human cell-culture models of pathogenic aggregation. A C. elegans strain expressing human Aβ1-42 in muscle, leading to AD-like amyloidopathy, developed fewer and smaller aggregates after PNR886 or PNR962 treatment. Moreover, age-progressive paralysis was reduced 90% by PNR886 and 75% by PNR962, and "healthspan" (the median duration of spontaneous motility) was extended 29% and 62%, respectively. These TDZD analogs also extended wild-type C. elegans lifespan by 15-30% (p < 0.001), placing them among the most effective life-extension drugs. Because the lead drug in this family, TDZD-8, inhibits GSK3β, we used molecular-dynamic tools to assess whether these analogs may also target GSK3β. In silico modeling predicted that PNR886 or PNR962 would bind to the same allosteric pocket of inactive GSK3β as TDZD-8, employing the same pharmacophore but attaching with greater avidity. PNR886 and PNR962 are thus compelling candidate drugs for treatment of tau- and amyloid-associated neurodegenerative diseases such as AD, potentially also reducing all-cause mortality.
Collapse
Affiliation(s)
- Samuel Kakraba
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Srinivas Ayyadevara
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Nirjal Mainali
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Suresh Bowroju
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Ramani Atluri
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Steven W. Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Robert J. Shmookler Reis
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| |
Collapse
|
25
|
Kulichikhin KY, Malikova OA, Zobnina AE, Zalutskaya NM, Rubel AA. Interaction of Proteins Involved in Neuronal Proteinopathies. Life (Basel) 2023; 13:1954. [PMID: 37895336 PMCID: PMC10608209 DOI: 10.3390/life13101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/04/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Proteinopathy is characterized by the accumulation of aggregates of a specific protein in a target organ, tissue, or cell. The aggregation of the same protein can cause different pathologies as single protein can adopt various amyloidogenic, disease-specific conformations. The conformation governs the interaction of amyloid aggregates with other proteins that are prone to misfolding and, thus, determines disease-specific spectrum of concomitant pathologies. In this regard, a detailed description of amyloid protein conformation as well as spectrum of its interaction with other proteins become a key point for drafting of precise description of the disease. The majority of clinical cases of neuronal proteinopathies is caused by the aggregation of rather limited range of amyloidogenic proteins. Here, we provided the characterization of pathologies, related to the aggregation of amyloid β peptide, tau protein, α-synuclein, TDP-43, and amylin, giving a short description of pathologies themselves, recent advances in elucidation of misfolded protein conformation, with emphasis on those protein aggregates extracted from biological samples, what is known about the interaction of this proteins, and the influence of this interaction on the progression of underlying disease and comorbidities.
Collapse
Affiliation(s)
- Konstantin Y. Kulichikhin
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Oksana A. Malikova
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Anastasia E. Zobnina
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| | - Natalia M. Zalutskaya
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, 192019 St. Petersburg, Russia;
| | - Aleksandr A. Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia; (O.A.M.); (A.E.Z.)
| |
Collapse
|
26
|
Kachkin DV, Lashkul VV, Gorsheneva NA, Fedotov SA, Rubel MS, Chernoff YO, Rubel AA. The Aβ42 Peptide and IAPP Physically Interact in a Yeast-Based Assay. Int J Mol Sci 2023; 24:14122. [PMID: 37762425 PMCID: PMC10531723 DOI: 10.3390/ijms241814122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Numerous studies have demonstrated that people with type 2 diabetes mellitus (associated with IAPP peptide aggregation) show an increased incidence of Alzheimer's disease (associated with Aβ aggregation), but the mechanism responsible for this correlation is presently unknown. Here, we applied a yeast-based model to study the interactions of IAPP with PrP (associated with TSEs) and with the Aβ42 peptide. We demonstrated that fluorescently tagged IAPP forms detergent-resistant aggregates in yeast cells. Using the FRET approach, we showed that IAPP and Aβ aggregates co-localize and physically interact in yeast cells. We also showed that this interaction is specific and that there is no interaction between IAPP and PrP in the yeast system. Our data confirmed a direct physical interaction between IAPP and Aβ42 aggregates in a living cell. Based on these findings, we hypothesize that this interaction may play a crucial role in seeding Aβ42 aggregation in T2DM patients, thereby promoting the development of AD.
Collapse
Affiliation(s)
- Daniel V. Kachkin
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia; (D.V.K.); (S.A.F.)
| | - Veronika V. Lashkul
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia; (D.V.K.); (S.A.F.)
| | - Natalia A. Gorsheneva
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia; (D.V.K.); (S.A.F.)
| | - Sergey A. Fedotov
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia; (D.V.K.); (S.A.F.)
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg 199034, Russia
| | - Maria S. Rubel
- Laboratory of DNA-Nanosensor Diagnostics, SCAMT Institute, ITMO University, St. Petersburg 191002, Russia;
| | - Yury O. Chernoff
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Aleksandr A. Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg 199034, Russia; (D.V.K.); (S.A.F.)
- Pediatric Research and Clinical Center for Infectious Diseases, Department of Medical Microbiology and Molecular Epidemiology, St. Petersburg 197022, Russia
| |
Collapse
|
27
|
Gutierrez-Merino C. Brain Hydrophobic Peptides Antagonists of Neurotoxic Amyloid β Peptide Monomers/Oligomers-Protein Interactions. Int J Mol Sci 2023; 24:13846. [PMID: 37762148 PMCID: PMC10531495 DOI: 10.3390/ijms241813846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Amyloid β (Aβ) oligomers have been linked to Alzheimer's disease (AD) pathogenesis and are the main neurotoxic forms of Aβ. This review focuses on the following: (i) the Aβ(1-42):calmodulin interface as a model for the design of antagonist Aβ peptides and its limitations; (ii) proteolytic degradation as the major source of highly hydrophobic peptides in brain cells; and (iii) brain peptides that have been experimentally demonstrated to bind to Aβ monomers or oligomers, Aβ fibrils, or Aβ plaques. It is highlighted that the hydrophobic amino acid residues of the COOH-terminal segment of Aβ(1-42) play a key role in its interaction with intracellular protein partners linked to its neurotoxicity. The major source of highly hydrophobic endogenous peptides of 8-10 amino acids in neurons is the proteasome activity. Many canonical antigen peptides bound to the major histocompatibility complex class 1 are of this type. These highly hydrophobic peptides bind to Aβ and are likely to be efficient antagonists of the binding of Aβ monomers/oligomers concentrations in the nanomolar range with intracellular proteins. Also, their complexation with Aβ will protect them against endopeptidases, suggesting a putative chaperon-like physiological function for Aβ that has been overlooked until now. Remarkably, the hydrophobic amino acid residues of Aβ responsible for the binding of several neuropeptides partially overlap with those playing a key role in its interaction with intracellular protein partners that mediates its neurotoxicity. Therefore, these latter neuropeptides are also potential candidates to antagonize Aβ peptides binding to target proteins. In conclusion, the analysis performed in this review points out that hydrophobic endogenous brain neuropeptides could be valuable biomarkers to evaluate the risk of the onset of sporadic AD, as well as for the prognosis of AD.
Collapse
Affiliation(s)
- Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
28
|
Kell DB, Pretorius E. Are fibrinaloid microclots a cause of autoimmunity in Long Covid and other post-infection diseases? Biochem J 2023; 480:1217-1240. [PMID: 37584410 DOI: 10.1042/bcj20230241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
It is now well established that the blood-clotting protein fibrinogen can polymerise into an anomalous form of fibrin that is amyloid in character; the resultant clots and microclots entrap many other molecules, stain with fluorogenic amyloid stains, are rather resistant to fibrinolysis, can block up microcapillaries, are implicated in a variety of diseases including Long COVID, and have been referred to as fibrinaloids. A necessary corollary of this anomalous polymerisation is the generation of novel epitopes in proteins that would normally be seen as 'self', and otherwise immunologically silent. The precise conformation of the resulting fibrinaloid clots (that, as with prions and classical amyloid proteins, can adopt multiple, stable conformations) must depend on the existing small molecules and metal ions that the fibrinogen may (and is some cases is known to) have bound before polymerisation. Any such novel epitopes, however, are likely to lead to the generation of autoantibodies. A convergent phenomenology, including distinct conformations and seeding of the anomalous form for initiation and propagation, is emerging to link knowledge in prions, prionoids, amyloids and now fibrinaloids. We here summarise the evidence for the above reasoning, which has substantial implications for our understanding of the genesis of autoimmunity (and the possible prevention thereof) based on the primary process of fibrinaloid formation.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
29
|
Toledo PL, Vazquez DS, Gianotti AR, Abate MB, Wegbrod C, Torkko JM, Solimena M, Ermácora MR. Condensation of the β-cell secretory granule luminal cargoes pro/insulin and ICA512 RESP18 homology domain. Protein Sci 2023; 32:e4649. [PMID: 37159024 PMCID: PMC10201709 DOI: 10.1002/pro.4649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/10/2023]
Abstract
ICA512/PTPRN is a receptor tyrosine-like phosphatase implicated in the biogenesis and turnover of the insulin secretory granules (SGs) in pancreatic islet beta cells. Previously we found biophysical evidence that its luminal RESP18 homology domain (RESP18HD) forms a biomolecular condensate and interacts with insulin in vitro at close-to-neutral pH, that is, in conditions resembling those present in the early secretory pathway. Here we provide further evidence for the relevance of these findings by showing that at pH 6.8 RESP18HD interacts also with proinsulin-the physiological insulin precursor found in the early secretory pathway and the major luminal cargo of β-cell nascent SGs. Our light scattering analyses indicate that RESP18HD and proinsulin, but also insulin, populate nanocondensates ranging in size from 15 to 300 nm and 10e2 to 10e6 molecules. Co-condensation of RESP18HD with proinsulin/insulin transforms the initial nanocondensates into microcondensates (size >1 μm). The intrinsic tendency of proinsulin to self-condensate implies that, in the ER, a chaperoning mechanism must arrest its spontaneous intermolecular condensation to allow for proper intramolecular folding. These data further suggest that proinsulin is an early driver of insulin SG biogenesis, in a process in which its co-condensation with RESP18HD participates in their phase separation from other secretory proteins in transit through the same compartments but destined to other routes. Through the cytosolic tail of ICA512, proinsulin co-condensation with RESP18HD may further orchestrate the recruitment of cytosolic factors involved in membrane budding and fission of transport vesicles and nascent SGs.
Collapse
Affiliation(s)
- Pamela L. Toledo
- Departamento de Ciencia y TecnologíaUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
- Grupo de Biología Estructural y Biotecnología, IMBICE, CONICETUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
| | - Diego S. Vazquez
- Departamento de Ciencia y TecnologíaUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
- Grupo de Biología Estructural y Biotecnología, IMBICE, CONICETUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
| | - Alejo R. Gianotti
- Departamento de Ciencia y TecnologíaUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
- Grupo de Biología Estructural y Biotecnología, IMBICE, CONICETUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
| | - Milagros B. Abate
- Departamento de Ciencia y TecnologíaUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
- Grupo de Biología Estructural y Biotecnología, IMBICE, CONICETUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
| | - Carolin Wegbrod
- Department of Molecular DiabetologyUniversity Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- German Center for Diabetes Research (DZD e.V.)NeuherbergGermany
| | - Juha M. Torkko
- Department of Molecular DiabetologyUniversity Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- German Center for Diabetes Research (DZD e.V.)NeuherbergGermany
| | - Michele Solimena
- Department of Molecular DiabetologyUniversity Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, TU DresdenDresdenGermany
- German Center for Diabetes Research (DZD e.V.)NeuherbergGermany
| | - Mario R. Ermácora
- Departamento de Ciencia y TecnologíaUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
- Grupo de Biología Estructural y Biotecnología, IMBICE, CONICETUniversidad Nacional de QuilmesProvincia de Buenos AiresArgentina
| |
Collapse
|
30
|
Cardinali DP, Garay A. Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder. Brain Sci 2023; 13:brainsci13050797. [PMID: 37239269 DOI: 10.3390/brainsci13050797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Dream-enactment behavior that emerges during episodes of rapid eye movement (REM) sleep without muscle atonia is a parasomnia known as REM sleep behavior disorder (RBD). RBD constitutes a prodromal marker of α-synucleinopathies and serves as one of the best biomarkers available to predict diseases such as Parkinson disease, multiple system atrophy and dementia with Lewy bodies. Most patients showing RBD will convert to an α-synucleinopathy about 10 years after diagnosis. The diagnostic advantage of RBD relies on the prolonged prodromal time, its predictive power and the absence of disease-related treatments that could act as confounders. Therefore, patients with RBD are candidates for neuroprotection trials that delay or prevent conversion to a pathology with abnormal α-synuclein metabolism. The administration of melatonin in doses exhibiting a chronobiotic/hypnotic effect (less than 10 mg daily) is commonly used as a first line treatment (together with clonazepam) of RBD. At a higher dose, melatonin may also be an effective cytoprotector to halt α-synucleinopathy progression. However, allometric conversion doses derived from animal studies (in the 100 mg/day range) are rarely employed clinically regardless of the demonstrated absence of toxicity of melatonin in phase 1 pharmacological studies with doses up to 100 mg in normal volunteers. This review discusses the application of melatonin in RBD: (a) as a symptomatic treatment in RBD; (b) as a possible disease-modifying treatment in α-synucleinopathies. To what degree melatonin has therapeutic efficacy in the prevention of α-synucleinopathies awaits further investigation, in particular multicenter double-blind trials.
Collapse
Affiliation(s)
- Daniel P Cardinali
- CENECON, Faculty of Medical Sciences, University of Buenos Aires, Buenos Aires C1431FWO, Argentina
| | - Arturo Garay
- Unidad de Medicina del Sueño-Sección Neurología, Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Buenos Aires C1431FWO, Argentina
| |
Collapse
|
31
|
Meng L, Li Y, Liu C, Zhang G, Chen J, Xiong M, Pan L, Zhang X, Chen G, Xiong J, Liu C, Xu X, Bu L, Zhang Z, Zhang Z. Islet Amyloid Polypeptide Triggers α-synuclein Pathology in Parkinson's Disease. Prog Neurobiol 2023; 226:102462. [PMID: 37150314 DOI: 10.1016/j.pneurobio.2023.102462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/28/2023] [Accepted: 05/01/2023] [Indexed: 05/09/2023]
Abstract
Pathologic aggregation and prion-like propagation of α-synuclein (α-syn) are the hallmarks of Parkinson's disease (PD). Emerging evidence shows that type 2 diabetes mellitus (T2DM) is a risk factor for PD. Interestingly, T2DM is characterized by the amyloid deposition of islet amyloid polypeptide (IAPP) in the pancreas. Although T2DM and PD share pathological similarities, the underlying molecular mechanisms bridging these two diseases remain unknown. Here, we report that IAPP co-deposits with α-syn in the brains of PD patients. IAPP interacts with α-syn and accelerates its aggregation. In addition, the IAPP-seeded α-syn fibrils show enhanced seeding activity and neurotoxicity compared with pure α-syn fibrils in vitro and in vivo. Strikingly, intravenous injection of IAPP fibrils into α-syn A53T transgenic mice or human SNCA transgenic mice accelerated the aggregation of α-syn and PD-like motor deficits. Taken together, these findings support that IAPP acts as a trigger of α-syn pathology in PD, and provide a mechanistic explanation for the increased risk and faster progression of PD in patients with T2DM.
Collapse
Affiliation(s)
- Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yiming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Congcong Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiehui Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guiqin Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chaoyang Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan 430073, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lihong Bu
- PET-CT/MRI Center, Faculty of Radiology and Nuclear Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
32
|
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
|
33
|
Maity D. Recent advances in the modulation of amyloid protein aggregation using the supramolecular host-guest approaches. Biophys Chem 2023; 297:107022. [PMID: 37058879 DOI: 10.1016/j.bpc.2023.107022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Misfolding of proteins is associated with many incurable diseases in human beings. Understanding the process of aggregation from monomers to fibrils, the characterization of all intermediate species, and the origin of toxicity is very challenging. Extensive research including computational and experimental shed some light on these tricky phenomena. Non-covalent interactions between amyloidogenic domains of proteins play a major role in their self-assembly which can be disrupted by designed chemical tools. This will lead to the development of inhibitors of detrimental amyloid formations. In supramolecular host-guest chemistry approaches, different macrocycles function as hosts for encapsulating hydrophobic guests, i.e. phenylalanine residues of proteins, in their hydrophobic cavities via non-covalent interactions. In this way, they can disrupt the interactions between adjacent amyloidogenic proteins and prevent their self-aggregation. This supramolecular approach has also emerged as a prospective tool to modify the aggregation of several amyloidogenic proteins. In this review, we discussed recent supramolecular host-guest chemistry-based strategies for the inhibition of amyloid protein aggregation.
Collapse
Affiliation(s)
- Debabrata Maity
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
34
|
PPARs and Their Neuroprotective Effects in Parkinson's Disease: A Novel Therapeutic Approach in α-Synucleinopathy? Int J Mol Sci 2023; 24:ijms24043264. [PMID: 36834679 PMCID: PMC9963164 DOI: 10.3390/ijms24043264] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is the most common α-synucleinopathy worldwide. The pathognomonic hallmark of PD is the misfolding and propagation of the α-synuclein (α-syn) protein, observed in post-mortem histopathology. It has been hypothesized that α-synucleinopathy triggers oxidative stress, mitochondrial dysfunction, neuroinflammation, and synaptic dysfunction, leading to neurodegeneration. To this date, there are no disease-modifying drugs that generate neuroprotection against these neuropathological events and especially against α-synucleinopathy. Growing evidence suggests that peroxisome proliferator-activated receptor (PPAR) agonists confer neuroprotective effects in PD, however, whether they also confer an anti-α-synucleinopathy effect is unknown. Here we analyze the reported therapeutic effects of PPARs, specifically the gamma isoform (PPARγ), in preclinical PD animal models and clinical trials for PD, and we suggest possible anti-α-synucleinopathy mechanisms acting downstream from these receptors. Elucidating the neuroprotective mechanisms of PPARs through preclinical models that mimic PD as closely as possible will facilitate the execution of better clinical trials for disease-modifying drugs in PD.
Collapse
|
35
|
Cullinane PW, de Pablo Fernandez E, König A, Outeiro TF, Jaunmuktane Z, Warner TT. Type 2 Diabetes and Parkinson's Disease: A Focused Review of Current Concepts. Mov Disord 2023; 38:162-177. [PMID: 36567671 DOI: 10.1002/mds.29298] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 12/27/2022] Open
Abstract
Highly reproducible epidemiological evidence shows that type 2 diabetes (T2D) increases the risk and rate of progression of Parkinson's disease (PD), and crucially, the repurposing of certain antidiabetic medications for the treatment of PD has shown early promise in clinical trials, suggesting that the effects of T2D on PD pathogenesis may be modifiable. The high prevalence of T2D means that a significant proportion of patients with PD may benefit from personalized antidiabetic treatment approaches that also confer neuroprotective benefits. Therefore, there is an immediate need to better understand the mechanistic relation between these conditions and the specific molecular pathways affected by T2D in the brain. Although there is considerable evidence that processes such as insulin signaling, mitochondrial function, autophagy, and inflammation are involved in the pathogenesis of both PD and T2D, the primary aim of this review is to highlight the evidence showing that T2D-associated dysregulation of these pathways occurs not only in the periphery but also in the brain and how this may facilitate neurodegeneration in PD. We also discuss the challenges involved in disentangling the complex relationship between T2D, insulin resistance, and PD, as well as important questions for further research. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eduardo de Pablo Fernandez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
36
|
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
|
37
|
Yang Y, Wang Q, Li G, Guo W, Yang Z, Liu H, Deng X. Cysteine-Derived Chiral Carbon Quantum Dots: A Fibrinolytic Activity Regulator for Plasmin to Target the Human Islet Amyloid Polypeptide for Type 2 Diabetes Mellitus. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2617-2629. [PMID: 36596222 DOI: 10.1021/acsami.2c17975] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The fibrillization and deposition of the human islet amyloid polypeptide (hIAPP) are the pathological hallmark of type 2 diabetes mellitus (T2DM), and these insoluble fibrotic depositions of hIAPP are considered to strongly affect insulin secretion by inducing toxicity toward pancreatic islet β-cells. The current strategy of preventing amyloid aggregation by nanoparticle-assisted inhibitors can only disassemble fibrotic amyloids into more toxic oligomers and/or protofibrils. Herein, for the first time, we propose a type of cysteine-derived chiral carbon quantum dot (CQD) that targets plasmin, a core natural fibrinolytic protease in humans. These CQDs can serve as fibrinolytic activity regulators for plasmin to cleave hIAPP into nontoxic polypeptides or into even smaller amino acid fragments, thus alleviating hIAPP's fibrotic amyloid-induced cytotoxicity. Our experiments indicate that chiral CQDs have opposing effects on plasmin activity. The l-CQDs promote the cleavage of hIAPP by enhancing plasmin activity at a promotion ratio of 23.2%, thus protecting β-cells from amyloid-induced toxicity. In contrast, the resultant d-CQDs significantly inhibit proteolysis, decreasing plasmin activity by 31.5% under the same reaction conditions. Second harmonic generation (SHG) microscopic imaging is initially used to dynamically characterize hIAPP before and after proteolysis. The l-CQD promotion of plasmin activity thus provides a promising avenue for the hIAPP-targeted treatment of T2DM to treat low fibrinolytic activity, while the d-CQDs, as inhibitors of plasmin activity, may improve patient survival for hyperfibrinolytic conditions, such as those existing during surgeries and traumas.
Collapse
Affiliation(s)
- Yongzhen Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Qin Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Gongjian Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Wenjing Guo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou510530, China
| | - Zuojun Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Hao Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou510631, China
- Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou510631, China
| |
Collapse
|
38
|
Ge WY, Deng X, Shi WP, Lin WJ, Chen LL, Liang H, Wang XT, Zhang TD, Zhao FZ, Guo WH, Yin DC. Amyloid Protein Cross-Seeding Provides a New Perspective on Multiple Diseases In Vivo. Biomacromolecules 2023; 24:1-18. [PMID: 36507729 DOI: 10.1021/acs.biomac.2c01233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Amyloid protein cross-seeding is a peculiar phenomenon of cross-spreading among different diseases. Unlike traditional infectious ones, diseases caused by amyloid protein cross-seeding are spread by misfolded proteins instead of pathogens. As a consequence of the interactions among misfolded heterologous proteins or polypeptides, amyloid protein cross-seeding is considered to be the crucial cause of overlapping pathological transmission between various protein misfolding disorders (PMDs) in multiple tissues and cells. Here, we briefly review the phenomenon of cross-seeding among amyloid proteins. As an interesting example worth mentioning, the potential links between the novel coronavirus pneumonia (COVID-19) and some neurodegenerative diseases might be related to the amyloid protein cross-seeding, thus may cause an undesirable trend in the incidence of PMDs around the world. We then summarize the theoretical models as well as the experimental techniques for studying amyloid protein cross-seeding. Finally, we conclude with an outlook on the challenges and opportunities for basic research in this field. Cross-seeding of amyloid opens up a new perspective in our understanding of the process of amyloidogenesis, which is crucial for the development of new treatments for diseases. It is therefore valuable but still challenging to explore the cross-seeding system of amyloid protein as well as to reveal the structural basis and the intricate processes.
Collapse
Affiliation(s)
- Wan-Yi Ge
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xudong Deng
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Wen-Pu Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Wen-Juan Lin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Liang-Liang Chen
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Huan Liang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xue-Ting Wang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tuo-Di Zhang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Feng-Zhu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.,Non-commissioned Officer School, Army Medical University, Shijiazhuang 050081, China
| | - Wei-Hong Guo
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
39
|
Park J, Choi S, Kim R. Association between prediabetes and cognitive function in Parkinson's disease. Brain Behav 2023; 13:e2838. [PMID: 36448303 PMCID: PMC9847602 DOI: 10.1002/brb3.2838] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION It remains largely unknown whether prediabetes is related to cognitive impairment in Parkinson's disease (PD). This study aimed to assess the association between prediabetes and cognitive function in PD patients. METHODS In this cross-sectional study, 262 PD patients (age, 69.8 ± 10.3 years; Hoehn-Yahr stage, 2.3 ± 0.8) were classified into diabetes (glycated hemoglobin [HbA1c] ≥6.5% or previously diagnosed, n = 76), prediabetes (5.7%-6.4%, n = 90), or diabetes free (≤5.6%, n = 96) groups. Cognitive function was measured using the Montreal Cognitive Assessment (MoCA) test. RESULTS Both the diabetes and prediabetes groups had significantly lower MoCA scores (17.0 ± 6.6 and 18.0 ± 6.1, respectively) than the diabetes free group (20.0 ± 5.7), even after adjusting for potential confounders (p = .002 and p = .008, respectively). In the combined group of prediabetes and diabetes free patients, higher HbA1c levels significantly correlated with lower MoCA scores (p = .031). There was a significant interaction of diabetes status with age, but not with the duration of PD, on cognitive function. CONCLUSION In addition to diabetes, prediabetes may negatively affect cognitive function in PD patients. Further prospective longitudinal studies are necessary to clarify the impact of prediabetes on the cognitive trajectory of these patients.
Collapse
Affiliation(s)
- Joah Park
- Department of Medicine, Inha University College of Medicine, Incheon, Korea
| | - Seohee Choi
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Ryul Kim
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
40
|
Yu H, Sun T, He X, Wang Z, Zhao K, An J, Wen L, Li JY, Li W, Feng J. Association between Parkinson's Disease and Diabetes Mellitus: From Epidemiology, Pathophysiology and Prevention to Treatment. Aging Dis 2022; 13:1591-1605. [PMID: 36465171 PMCID: PMC9662283 DOI: 10.14336/ad.2022.0325] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/25/2022] [Indexed: 08/27/2023] Open
Abstract
Diabetes mellitus (DM) and Parkinson's disease (PD) are both age-related diseases of global concern being among the most common chronic metabolic and neurodegenerative diseases, respectively. While both diseases can be genetically inherited, environmental factors play a vital role in their pathogenesis. Moreover, DM and PD have common underlying molecular mechanisms, such as misfolded protein aggregation, mitochondrial dysfunction, oxidative stress, chronic inflammation, and microbial dysbiosis. Recently, epidemiological and experimental studies have reported that DM affects the incidence and progression of PD. Moreover, certain antidiabetic drugs have been proven to decrease the risk of PD and delay its progression. In this review, we elucidate the epidemiological and pathophysiological association between DM and PD and summarize the antidiabetic drugs used in animal models and clinical trials of PD, which may provide reference for the clinical translation of antidiabetic drugs in PD treatment.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhen Wang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Kaidong Zhao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Jing An
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jia-Yi Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Wen Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
41
|
Crossroads between copper ions and amyloid formation in Parkinson's disease. Essays Biochem 2022; 66:977-986. [PMID: 35757906 PMCID: PMC9760422 DOI: 10.1042/ebc20220043] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 12/25/2022]
Abstract
Copper (Cu) ion dys-homeostasis and α-synclein amyloid deposits are two hallmarks of Parkinson's disease (PD). Here, I will discuss the connections between these features, with a major focus on the role of Cu in the α-synuclein (aS) amyloid formation process. The structurally disordered aS monomer can bind to both redox states of Cu (i.e., oxidized Cu(II) and reduced Cu(I)) with high affinity in vitro. Notably, the presence of Cu(II) (in absence of aS N-terminal acetylation) and Cu(I) (when in complex with the copper chaperone Atox1) modulate aS assembly into β-structured amyloids in opposite directions in vitro. Albeit the link to biological relevance is not fully unraveled, existing observations clearly emphasize the need for more knowledge on this interplay and its consequences to eventually combat destructive reactions that promote PD.
Collapse
|
42
|
Liu C, Liu Z, Fang Y, Du Z, Yan Z, Yuan X, Dai L, Yu T, Xiong M, Tian Y, Li H, Li F, Zhang J, Meng L, Wang Z, Jiang H, Zhang Z. Exposure to the environmentally toxic pesticide maneb induces Parkinson's disease-like neurotoxicity in mice: A combined proteomic and metabolomic analysis. CHEMOSPHERE 2022; 308:136344. [PMID: 36087732 DOI: 10.1016/j.chemosphere.2022.136344] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/03/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Maneb is a typical dithiocarbamate fungicide that has been extensively used worldwide. Epidemiological evidence shows that exposure to maneb is an environmental risk factor for Parkinson's disease (PD). However, the mechanisms underlying maneb-induced neurotoxicity have yet to be elucidated. In this study, we exposed SH-SY5Y cells to maneb at environmentally relevant concentrations (0, 0.1, 5, 10 mg/L) and found that maneb dose-dependently decreased the cell viability. Furthermore, maneb (60 mg/kg) induced PD-like motor impairment in α-synuclein A53T transgenic mice. The results of tandem mass tag (TMT) proteomics and metabolomics studies of mouse brain and serum revealed significant changes in proteins and metabolites in the pathways involved in the neurotransmitter system. The omics results were verified by targeted metabolomics and Western blot analysis, which demonstrated that maneb induced disturbance of the PD-related pathways, including the phenylalanine and tryptophan metabolism pathways, dopaminergic synapse, synaptic vesicle cycle, mitochondrial dysfunction, and oxidative stress. In addition, the PD-like phenotype induced by maneb was attenuated by the asparagine endopeptidase (AEP) inhibitor compound #11 (CP11) (10 mg/kg), indicating that AEP may play a role in maneb-induced neurotoxicity. To the best of our knowledge, this is the first study to investigate the molecular mechanisms underlying maneb-induced PD-like phenotypes using multiomics analysis, which identified novel therapeutic targets for PD associated with pesticides and other environmental pollutants.
Collapse
Affiliation(s)
- Chaoyang Liu
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zehua Liu
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Yanyan Fang
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Zhen Du
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Zhi Yan
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Xin Yuan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Honghu Li
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Fei Li
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Jingdong Zhang
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China; Department of Environmental Engineering, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhihao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiqiang Jiang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
43
|
Faizan M, Sarkar A, Singh MP. Type 2 diabetes mellitus augments Parkinson's disease risk or the other way around: Facts, challenges and future possibilities. Ageing Res Rev 2022; 81:101727. [PMID: 36038113 DOI: 10.1016/j.arr.2022.101727] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 08/01/2022] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
About 10% of the adult population is living with type 2 diabetes mellitus (T2DM) and 1% of the population over 60 years of age is suffering from Parkinson's disease (PD). A school of thought firmly believes that T2DM, an age-related disease, augments PD risk. Such relationship is reflected from the severity of PD symptoms in drug naive subjects possessing T2DM. Onset of Parkinsonian feature in case controls possessing T2DM corroborates the role of hyperglycemia in PD. A few cohort, meta-analysis and animal studies have shown an increased PD risk owing to insulin resistance. High fat diet and role of insulin signaling in the regulation of sugar metabolism, oxidative stress, α-synuclein aggregation and accumulation, inflammatory response and mitochondrial function in PD models and sporadic PD further connect the two. Although little is reported about the implication of PD in hyperglycemia and T2DM, a few studies have also contradicted. Ameliorative effect of anti-diabetic drugs on Parkinsonian symptoms and vague outcome of anti-PD medications in T2DM patients also suggest a link. The article reviews the literature supporting augmented risk of one by the other, analysis of proof of the concept, facts, challenges, future possibilities and standpoint on the subject.
Collapse
Affiliation(s)
- Mohd Faizan
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India
| | - Alika Sarkar
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India.
| |
Collapse
|
44
|
Guillemain G, Lacapere JJ, Khemtemourian L. Targeting hIAPP fibrillation: A new paradigm to prevent β-cell death? BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184002. [PMID: 35868406 DOI: 10.1016/j.bbamem.2022.184002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Loss of pancreatic β-cell mass is deleterious for type 2 diabetes patients since it reduces insulin production, critical for glucose homeostasis. The main research axis developed over the last few years was to generate new pancreatic β-cells or to transplant pancreatic islets as occurring for some specific type 1 diabetes patients. We evaluate here a new paradigm consisting in preservation of β-cells by prevention of human islet amyloid polypeptide (hIAPP) oligomers and fibrils formation leading to pancreatic β-cell death. We review the hIAPP physiology and the pathology that contributes to β-cell destruction, deciphering the various cellular steps that could be involved. Recent progress in understanding other amyloidosis such as Aβ, Tau, α-synuclein or prion, involved in neurodegenerative processes linked with inflammation, has opened new research lines of investigations to preserve neuronal cells. We evaluate and estimate their transposition to the pancreatic β-cells preservation. Among them is the control of reactive oxygen species (ROS) production occurring with inflammation and the possible implication of the mitochondrial translocator protein as a diagnostic and therapeutic target. The present review also focuses on other amyloid forming proteins from molecular to physiological and physiopathological points of view that could help to better decipher hIAPP-induced β-cell death mechanisms and to prevent hIAPP fibril formation.
Collapse
Affiliation(s)
- Ghislaine Guillemain
- Sorbonne Université, Institut Hospitalo-Universitaire, Inserm UMR_S938, Institute of Cardio metabolism and Nutrition (ICAN), Centre de recherche de St-Antoine (CRSA), 27 rue de Chaligny, F-75012 Paris, France.
| | - Jean-Jacques Lacapere
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS UMR 7203, Laboratoire des BioMolécules (LBM), 4 place Jussieu, F-75005 Paris, France.
| | - Lucie Khemtemourian
- CBMN, CNRS UMR 5248, IPB, Univ. Bordeaux, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France.
| |
Collapse
|
45
|
Akbarian M, Bahmani M, Chen SH, Yousefi R, Mohammadi-Samani S, Tayebi L, Panahi F, Farjadian F. Mechanisms behind the Fibrillation and Toxicity of Insulin Fibrils on Neuron Cells by Engineered Curcumin Analogs. ACS Chem Neurosci 2022; 13:2613-2631. [PMID: 35969719 DOI: 10.1021/acschemneuro.2c00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Among foods, the use of plant derivatives as promising drugs and/or excipients has been considered from various perspectives. In the present study, curcumin, which is one of the most important plant derivatives for biological uses, and four curcumin-based pyrido[2,3-d]pyrimidine analogs (C2-C5) were used for investigating the mechanism of insulin fibrillation and evaluating the cytotoxicity of insulin fibrils. The synthesized analogs differed in terms of hydrophobicity and electrostatic charge. The analogs with more hydrophobicity (C1 and C4) in both acidic and neutral environments were able to reduce the rate of insulin fibrillation and the degree of cross-linking in the produced fibrils. Additionally, the toxicity of these fibrils for neural cells (N2a cell line) was very low. However, they did not show any significant effects on the toxicity of non-neural cells (HEK293 cell line), indicating the effect of the biochemical surface diversity on determining the vulnerability to fibrils and even the mechanism of action of additives on cell line survival. Although negatively charged analogs were able to reduce insulin fibrillation in the acidic environment, they indicated an opposite effect in the neutral environment. The resultant fibrils in the acidic medium appeared with a well-distinguished filament, but they were very close at neutral pH levels. Moreover, such fibrils indicated very poor toxicity against the N2a cell line and had no significant effects on HEK293 cells. Considering the docking studies, by creatively using the size exclusion chromatography, it was suggested that analogs C2 and C3 were capable of binding to the C-terminal end of the insulin B chain (low affinity) and HisB10 (high affinity). Hence, it was suggested that different compounds could play different protecting and/or destroying roles in cell toxicity by blocking some ligands at the surface of neuron cells.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
- Department of Chemistry, National Cheng Kung University, Tainan701, Taiwan
| | - Marzieh Bahmani
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
- Department of Science, Medicine and Health, School of Chemistry and Molecular Bioscience, University of Wollongong, NSW, Wollongong2522, Australia
| | - Shu-Hui Chen
- Department of Chemistry, National Cheng Kung University, Tainan701, Taiwan
| | - Reza Yousefi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran1417466191, Iran
- Protein Chemistry Laboratory, Department of Biology, College of Sciences, Shiraz University, Shiraz7193371, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, Wisconsin53233-2186, United States
| | - Farhad Panahi
- Institut für Organische Chemie, Albert-Ludwigs-Universität Freiburg, Albertstraße 21, 79104 Freiburg im Breisgau, Germany
| | - Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz7146864685, Iran
| |
Collapse
|
46
|
Athauda D, Evans J, Wernick A, Virdi G, Choi ML, Lawton M, Vijiaratnam N, Girges C, Ben‐Shlomo Y, Ismail K, Morris H, Grosset D, Foltynie T, Gandhi S. The Impact of Type 2 Diabetes in Parkinson's Disease. Mov Disord 2022; 37:1612-1623. [PMID: 35699244 PMCID: PMC9543753 DOI: 10.1002/mds.29122] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2DM) is an established risk factor for developing Parkinson's disease (PD), but its effect on disease progression is not well understood. OBJECTIVE The aim of this study was to investigate the influence of T2DM on aspects of disease progression in PD. METHODS We analyzed data from the Tracking Parkinson's study to examine the effects of comorbid T2DM on PD progression and quality of life by comparing symptom severity scores assessing a range of motor and nonmotor symptoms. RESULTS We identified 167 (8.7%) patients with PD and T2DM (PD + T2DM) and 1763 (91.3%) patients with PD without T2DM (PD). After controlling for confounders, patients with T2DM had more severe motor symptoms, as assessed by Movement Disorder Society Unified Parkinson's Disease Rating Scale, Part III (25.8 [0.9] vs. 22.5 [0.3] P = 0.002), and nonmotor symptoms, as assessed by Non-Motor Symptoms Scale total (38.4 [2.5] vs. 31.8 [0.7] P < 0.001), and were significantly more likely to report loss of independence (odds ratio, 2.08; 95% confidence interval [CI]: 1.34-3.25; P = 0.001) and depression (odds ratio, 1.62; CI: 1.10-2.39; P = 0.015). Furthermore, over time, patients with T2DM had significantly faster motor symptom progression (P = 0.012), developed worse mood symptoms (P = 0.041), and were more likely to develop substantial gait impairment (hazard ratio, 1.55; CI: 1.07-2.23; P = 0.020) and mild cognitive impairment (hazard ratio, 1.7; CI: 1.24-2.51; P = 0.002) compared with the PD group. CONCLUSIONS In the largest study to date, T2DM is associated with faster disease progression in Parkinson's, highlighting an interaction between these two diseases. Because it is a potentially modifiable metabolic state, with multiple peripheral and central targets for intervention, it may represent a target for alleviating parkinsonian symptoms and slowing progression to disability and dementia. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dilan Athauda
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom,Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - James Evans
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Anna Wernick
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Gurvir Virdi
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Minee L. Choi
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Michael Lawton
- School of Social and Community MedicineUniversity of BristolBristolUnited Kingdom
| | - Nirosen Vijiaratnam
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Christine Girges
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Yoav Ben‐Shlomo
- School of Social and Community MedicineUniversity of BristolBristolUnited Kingdom
| | - Khalida Ismail
- Department of Psychological MedicineKing's College LondonUnited Kingdom
| | - Huw Morris
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Donald Grosset
- Institute of Neurological SciencesQueen Elizabeth University HospitalGlasgowUnited Kingdom
| | - Thomas Foltynie
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Sonia Gandhi
- Neurodegeneration Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom,UCL Queen Square Institute of NeurologyLondonUnited Kingdom,Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| |
Collapse
|
47
|
Abstract
Amyloids are organized suprastructural polypeptide arrangements. The prevalence of amyloid-related processes of pathophysiological relevance has been linked to aging-related degenerative diseases. Besides the role of genetic polymorphisms on the relative risk of amyloid diseases, the contributions of nongenetic ontogenic cluster of factors remain elusive. In recent decades, mounting evidences have been suggesting the role of essential micronutrients, in particular transition metals, in the regulation of amyloidogenic processes, both directly (such as binding to amyloid proteins) or indirectly (such as regulating regulatory partners, processing enzymes, and membrane transporters). The features of transition metals as regulatory cofactors of amyloid proteins and the consequences of metal dyshomeostasis in triggering amyloidogenic processes, as well as the evidences showing amelioration of symptoms by dietary supplementation, suggest an exaptative role of metals in regulating amyloid pathways. The self- and cross-talk replicative nature of these amyloid processes along with their systemic distribution support the concept of their metastatic nature. The role of amyloidosis as nutrient sensors would act as intra- and transgenerational epigenetic metabolic programming factors determining health span and life span, viability, which could participate as an evolutive selective pressure.
Collapse
Affiliation(s)
- Luís Maurício T R Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory for Macromolecules (LAMAC-DIMAV), National Institute of Metrology, Quality and Technology - INMETRO, Duque de Caxias, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Linking hIAPP misfolding and aggregation with type 2 diabetes mellitus: a structural perspective. Biosci Rep 2022; 42:231205. [PMID: 35475576 PMCID: PMC9118370 DOI: 10.1042/bsr20211297] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
There are over 40 identified human disorders that involve certain proteins folding incorrectly, accumulating in the body causing damage to cells and organs and causing disease. Type 2 Diabetes Mellitus (T2DM) is one of these protein misfolding disorders (PMDs) and involves human islet amyloid polypeptide (hIAPP) misfolding and accumulating in parts of the body, primarily in the pancreas, causing damage to islet cells and affecting glucose regulation. In this review, we have summarised our current understanding of what causes hIAPP to misfold, what conformations are found in different parts of the body with a particular focus on what is known about the structure of hIAPP and how this links to T2DM. Understanding the molecular basis behind these misfolding events is essential for understanding the role of hIAPP to develop better therapeutics since type 2 diabetes currently affects over 4.9 million people in the United Kingdom alone and is predicted to increase as our population ages.
Collapse
|
49
|
Al Adem K, Shanti A, Srivastava A, Homouz D, Thomas SA, Khair M, Stefanini C, Chan V, Kim TY, Lee S. Linking Alzheimer’s Disease and Type 2 Diabetes: Characterization and Inhibition of Cytotoxic Aβ and IAPP Hetero-Aggregates. Front Mol Biosci 2022; 9:842582. [PMID: 35372522 PMCID: PMC8968156 DOI: 10.3389/fmolb.2022.842582] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic self-aggregation of β-amyloid (Aβ) peptide and islet amyloid polypeptide (IAPP) is implicated in the pathogenesis of Alzheimer’s disease (AD) and Type 2 diabetes (T2D), respectively. Increasing evidence, particularly the co-deposition of Aβ and IAPP in both brain and pancreatic tissues, suggests that Aβ and IAPP cross-interaction may be responsible for a pathological link between AD and T2D. Here, we examined the nature of IAPP-Aβ40 co-aggregation and its inhibition by small molecules. In specific, we characterized the kinetic profiles, morphologies, secondary structures and toxicities of IAPP-Aβ40 hetero-assemblies and compared them to those formed by their homo-assemblies. We demonstrated that monomeric IAPP and Aβ40 form stable hetero-dimers and hetero-assemblies that further aggregate into β-sheet-rich hetero-aggregates that are toxic (cell viability <50%) to both PC-12 cells, a neuronal cell model, and RIN-m5F cells, a pancreatic cell model for β-cells. We then selected polyphenolic candidates to inhibit IAPP or Aβ40 self-aggregation and examined the inhibitory effect of the most potent candidate on IAPP-Aβ40 co-aggregation. We demonstrated that epigallocatechin gallate (EGCG) form inter-molecular hydrogen bonds with each of IAPP and Aβ40. We also showed that EGCG reduced hetero-aggregate formation and resulted in lower β-sheets content and higher unordered structures in IAPP-Aβ40-EGCG samples. Importantly, we showed that EGCG is highly effective in reducing the toxicity of IAPP-Aβ40 hetero-aggregates on both cell models, specifically at concentrations that are equivalent to or are 2.5-fold higher than the mixed peptide concentrations. To the best of our knowledge, this is the first study to report the inhibition of IAPP-Aβ40 co-aggregation by small molecules. We conclude that EGCG is a promising candidate to prevent co-aggregation and cytotoxicity of IAPP-Aβ40, which in turn, contribute to the pathological link between AD and T2D.
Collapse
Affiliation(s)
- Kenana Al Adem
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Aya Shanti
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Amit Srivastava
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dirar Homouz
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Physics, University of Houston, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Sneha Ann Thomas
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Tae-Yeon Kim
- Department of Civil Infrastructure and Environmental Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Sungmun Lee
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Khalifa University’s Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Sungmun Lee,
| |
Collapse
|
50
|
Orr AA, Kuhlmann SK, Tamamis P. Computational design of a β-wrapin's N-terminal domain with canonical and non-canonical amino acid modifications mimicking curcumin's proposed inhibitory function. Biophys Chem 2022; 286:106805. [DOI: 10.1016/j.bpc.2022.106805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
|