1
|
Augusto-Oliveira M, Arrifano GDP, Leal-Nazaré CG, Chaves-Filho A, Santos-Sacramento L, Lopes-Araujo A, Tremblay MÈ, Crespo-Lopez ME. Morphological diversity of microglia: Implications for learning, environmental adaptation, ageing, sex differences and neuropathology. Neurosci Biobehav Rev 2025; 172:106091. [PMID: 40049541 DOI: 10.1016/j.neubiorev.2025.106091] [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: 11/06/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Microglia are the brain resident macrophages that respond rapidly to any insult. These non-neuroectodermal cells are decorated with plenty of receptors allowing them to recognise and respond precisely to a multitude of stimuli. To do so, microglia undergo structural and functional changes aiming to actively keep the brain's homeostasis. However, some microglial responses, when sustained or exacerbated, can contribute to neuropathology and neurodegeneration. Many microglial molecular and cellular changes were identified that display a strong correlation with neuronal damage and neuroinflammation/disease status, as well as present key sex-related differences that modulate microglial outcomes. Nevertheless, the relationship between microglial structural and functional features is just beginning to be unravelled. Several reports show that microglia undergo soma and branch remodelling in response to environmental stimuli, ageing, neurodegenerative diseases, trauma, and systemic inflammation, suggesting a complex form and function link. Also, it is reasonable overall to suppose that microglia diminishing their process length and ramification also reduce their monitoring activity of synapses, which is critical for detecting any synaptic disturbance and performing synaptic remodelling. Elucidating the complex interactions between microglial morphological plasticity and its functional implications appears essential for the understanding of complex cognitive and behavioural processes in health and neuropathological conditions.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Amanda Lopes-Araujo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec, Qubec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada; College Member of the Royal Society of Canada, Canada.
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| |
Collapse
|
2
|
Ma H, Wang Z, Yu M, Zhai Y, Yan J. Aberrations in peripheral B lymphocytes and B lymphocyte subsets levels in Parkinson disease: a systematic review. Front Immunol 2025; 16:1526095. [PMID: 40230858 PMCID: PMC11994702 DOI: 10.3389/fimmu.2025.1526095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Objective The association of B lymphocytes and B lymphocyte subsets and Parkinson's disease (PD) is increasingly acknowledged. However, there is inconsistence in the alterations of B lymphocytes or B lymphocyte subsets in peripheral blood of PD patients. To comprehensively understand its changes in PD patients,it is necessary to conduct a systematic review on this subject. Methods PubMed, Cochrane Library, and MEDLINE databases were searched until 3rd February 2024. Results We included 20 studies (n=2658) to conduct this systematic review. We conducted a qualitative analysis to assess the alterations of B lymphocytes and B lymphocyte subsets in the peripheral blood of individuals with PD. And studies reviewed demonstrated a significant decrease in the number of B cells, as well as immune dysregulation in the B lymphocyte subsets of these patients' peripheral blood. Conclusion Studies reviewed demonstrated that PD is linked to abnormalities in B lymphocytes and/or B lymphocytes subsets in peripheral blood. This study provides a novel perspective into the pathogenesis of PD, and future investigations into the B lymphocytes and/or B lymphocyte subsets as biomarkers and therapeutic targets for PD is warranted.
Collapse
Affiliation(s)
- Hongxia Ma
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ziyuan Wang
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Miao Yu
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yibo Zhai
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Key laboratory of Neuromolecular Biology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
3
|
Park H, Kam TI, Dawson VL, Dawson TM. α-Synuclein pathology as a target in neurodegenerative diseases. Nat Rev Neurol 2025; 21:32-47. [PMID: 39609631 DOI: 10.1038/s41582-024-01043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
α-Synuclein misfolds into pathological forms that lead to various neurodegenerative diseases known collectively as α-synucleinopathies. In this Review, we provide a comprehensive overview of pivotal advances in α-synuclein research. We examine structural features and physiological functions of α-synuclein and summarize current insights into key post-translational modifications, such as nitration, phosphorylation, ubiquitination, sumoylation and truncation, considering their contributions to neurodegeneration. We also highlight the existence of disease-specific α-synuclein strains and their mechanisms of pathological spread, and discuss seed amplification assays and PET tracers as emerging diagnostic tools for detecting pathological α-synuclein in clinical settings. We also discuss α-synuclein aggregation and clearance mechanisms, and review cell-autonomous and non-cell-autonomous processes that contribute to neuronal death, including the roles of adaptive and innate immunity in α-synuclein-driven neurodegeneration. Finally, we highlight promising therapeutic approaches that target pathological α-synuclein and provide insights into emerging areas of research.
Collapse
Affiliation(s)
- Hyejin Park
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Adrienne Helis Malvin and Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [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: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
5
|
Bastioli G, Piccirillo S, Graciotti L, Carone M, Sprega G, Taoussi O, Preziuso A, Castaldo P. Calcium Deregulation in Neurodegeneration and Neuroinflammation in Parkinson's Disease: Role of Calcium-Storing Organelles and Sodium-Calcium Exchanger. Cells 2024; 13:1301. [PMID: 39120330 PMCID: PMC11311461 DOI: 10.3390/cells13151301] [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: 06/13/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that lacks effective treatment strategies to halt or delay its progression. The homeostasis of Ca2+ ions is crucial for ensuring optimal cellular functions and survival, especially for neuronal cells. In the context of PD, the systems regulating cellular Ca2+ are compromised, leading to Ca2+-dependent synaptic dysfunction, impaired neuronal plasticity, and ultimately, neuronal loss. Recent research efforts directed toward understanding the pathology of PD have yielded significant insights, particularly highlighting the close relationship between Ca2+ dysregulation, neuroinflammation, and neurodegeneration. However, the precise mechanisms driving the selective loss of dopaminergic neurons in PD remain elusive. The disruption of Ca2+ homeostasis is a key factor, engaging various neurodegenerative and neuroinflammatory pathways and affecting intracellular organelles that store Ca2+. Specifically, impaired functioning of mitochondria, lysosomes, and the endoplasmic reticulum (ER) in Ca2+ metabolism is believed to contribute to the disease's pathophysiology. The Na+-Ca2+ exchanger (NCX) is considered an important key regulator of Ca2+ homeostasis in various cell types, including neurons, astrocytes, and microglia. Alterations in NCX activity are associated with neurodegenerative processes in different models of PD. In this review, we will explore the role of Ca2+ dysregulation and neuroinflammation as primary drivers of PD-related neurodegeneration, with an emphasis on the pivotal role of NCX in the pathology of PD. Consequently, NCXs and their interplay with intracellular organelles may emerge as potentially pivotal players in the mechanisms underlying PD neurodegeneration, providing a promising avenue for therapeutic intervention aimed at halting neurodegeneration.
Collapse
Affiliation(s)
- Guendalina Bastioli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Marianna Carone
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
- Institute of Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, 8092 Zürich, Switzerland
| | - Giorgia Sprega
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Omayema Taoussi
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Pasqualina Castaldo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| |
Collapse
|
6
|
Marques CR, Campos J, Sampaio-Marques B, Antunes FF, Dos Santos Cunha RM, Silva D, Barata-Antunes S, Lima R, Fernandes-Platzgummer A, da Silva CL, Sousa RA, Salgado AJ. Secretome of bone marrow mesenchymal stromal cells cultured in a dynamic system induces neuroprotection and modulates microglial responsiveness in an α-synuclein overexpression rat model. Cytotherapy 2024; 26:700-713. [PMID: 38483360 DOI: 10.1016/j.jcyt.2024.02.008] [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: 10/16/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND AIMS Parkinson's disease (PD) is the second most common neurodegenerative disorder. The etiology of the disease remains largely unknown, but evidence have suggested that the overexpression and aggregation of alpha-synuclein (α-syn) play key roles in the pathogenesis and progression of PD. Mesenchymal stromal cells (MSCs) have been earning attention in this field, mainly due to their paracrine capacity. The bioactive molecules secreted by MSCs, i.e. their secretome, have been associated with enhanced neuronal survival as well as a strong modulatory capacity of the microenvironments where the disease develops. The selection of the appropriate animal model is crucial in studies of efficacy assessment. Given the involvement of α-syn in the pathogenesis of PD, the evidence generated from the use of animal models that develop a pathologic phenotype due to the action of this protein is extremely valuable. Therefore, in this work, we established an animal model based on the viral vector-mediated overexpression of A53T α-syn and studied the impact of the secretome of bone marrow mesenchymal stromal cells MSC(M) as a therapeutic strategy. METHODS Adult male rats were subjected to α-syn over expression in the nigrostriatal pathway to model dopaminergic neurodegeneration. The impact of locally administered secretome treatment from MSC(M) was studied. Motor impairments were assessed throughout the study coupled with whole-region (striatum and substantia nigra) confocal microscopy evaluation of histopathological changes associated with dopaminergic neurodegeneration and glial cell reactivity. RESULTS Ten weeks after lesion induction, the animals received secretome injections in the substantia nigra pars compacta (SNpc) and striatum (STR). The secretome used was produced from bone marrow mesenchymal stromal cells MSC(M) expanded in a spinner flask (SP) system. Nine weeks later, animals that received the viral vector containing the gene for A53T α-syn and treated with vehicle (Neurobasal-A medium) presented dopaminergic cell loss in the SNpc and denervation in the STR. The treatment with secretome significantly reduced the levels of α-syn in the SNpc and protected the dopaminergic neurons (DAn) within the SNpc and STR. CONCLUSIONS Our results are aligned with previous studies in both α-syn Caenorhabditis elegans models, as well as 6-OHDA rodent model, revealing that secretome exerted a neuroprotective effect. Moreover, these effects were associated with a modulation of microglial reactivity supporting an immunomodulatory role for the factors contained within the secretome. This further supports the development of new studies exploring the effects and the mechanism of action of secretome from MSC(M) against α-syn-induced neurotoxicity.
Collapse
Affiliation(s)
- Cláudia Raquel Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Filipa Ferreira Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Raquel Medina Dos Santos Cunha
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Deolinda Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Amandi Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa S.A., Barco, Portugal
| | - António José Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS-3Bs PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
7
|
Balzano T, Del Rey NLG, Esteban-García N, Reinares-Sebastián A, Pineda-Pardo JA, Trigo-Damas I, Obeso JA, Blesa J. Neurovascular and immune factors of vulnerability of substantia nigra dopaminergic neurons in non-human primates. NPJ Parkinsons Dis 2024; 10:118. [PMID: 38886348 PMCID: PMC11183116 DOI: 10.1038/s41531-024-00735-w] [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: 11/20/2023] [Accepted: 05/30/2024] [Indexed: 06/20/2024] Open
Abstract
Dopaminergic neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease (PD), while those in the dorsal tier and ventral tegmental area are relatively spared. The factors determining why these neurons are more vulnerable than others are still unrevealed. Neuroinflammation and immune cell infiltration have been demonstrated to be a key feature of neurodegeneration in PD. However, the link between selective dopaminergic neuron vulnerability, glial and immune cell response, and vascularization and their interactions has not been deciphered. We aimed to investigate the contribution of glial cell activation and immune cell infiltration in the selective vulnerability of ventral dopaminergic neurons within the midbrain in a non-human primate model of PD. Structural characteristics of the vasculature within specific regions of the midbrain were also evaluated. Parkinsonian monkeys exhibited significant microglial and astroglial activation in the whole midbrain, but no major sub-regional differences were observed. Remarkably, the ventral substantia nigra was found to be typically more vascularized compared to other regions. This feature might play some role in making this region more susceptible to immune cell infiltration under pathological conditions, as greater infiltration of both T- and B- lymphocytes was observed in parkinsonian monkeys. Higher vascular density within the ventral region of the SNc may be a relevant factor for differential vulnerability of dopaminergic neurons in the midbrain. The increased infiltration of T- and B- cells in this region, alongside other molecules or toxins, may also contribute to the susceptibility of dopaminergic neurons in PD.
Collapse
Affiliation(s)
- Tiziano Balzano
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Natalia López-González Del Rey
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Noelia Esteban-García
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Alejandro Reinares-Sebastián
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Inés Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain
| | - José A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain.
| |
Collapse
|
8
|
Garcia R, Zarate S, Srinivasan R. The Role of Astrocytes in Parkinson's Disease : Astrocytes in Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2024; 39:319-343. [PMID: 39190081 DOI: 10.1007/978-3-031-64839-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with a complex and multifactorial pathogenesis. This chapter delves into the critical role of astrocytes in PD. Once viewed as supporting cells in the central nervous system, astrocytes have emerged as key players in both maintaining neuronal health and contributing to neurodegeneration in PD. Their functions play a dual role in the progression of PD, ranging from protective functions like secretion of neurotrophic factors and clearance of α-synuclein to detrimental functions like promotion of neuroinflammation. This chapter is structured into three primary sections: the morphological and functional organization of astrocytes, astrocytic calcium signaling, and the role of astrocyte heterogeneity in PD. We provide a detailed exploration of astrocytic organelles, bidirectional astrocyte-neuron interactions, and the impact of astrocytic secretions such as antioxidant molecules and neurotrophic factors. Furthermore, we discuss the influence of astrocytes on non-neuronal cells, including interactions with microglia and the blood-brain barrier (BBB). By examining the multifaceted roles of astrocytes, in this chapter, we aim to bridge basic astrocyte biology with the clinical complexities of PD, offering insights into novel therapeutic strategies. The inclusion of astrocyte biology in our broader research approach will aid in the development of more effective treatment strategies for PD.
Collapse
Affiliation(s)
- Roger Garcia
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Sara Zarate
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
| |
Collapse
|
9
|
Ferreira SA, Li C, Klæstrup IH, Vitic Z, Rasmussen RK, Kirkegaard A, Toft GU, Betzer C, Svendsen P, Jensen PH, Luo Y, Etzerodt A, Moestrup SK, Romero-Ramos M. Sex-dimorphic neuroprotective effect of CD163 in an α-synuclein mouse model of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:164. [PMID: 38092806 PMCID: PMC10719342 DOI: 10.1038/s41531-023-00606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Alpha-synuclein (α-syn) aggregation and immune activation represent hallmark pathological events in Parkinson's disease (PD). The PD-associated immune response encompasses both brain and peripheral immune cells, although little is known about the immune proteins relevant for such a response. We propose that the upregulation of CD163 observed in blood monocytes and in the responsive microglia in PD patients is a protective mechanism in the disease. To investigate this, we used the PD model based on intrastriatal injections of murine α-syn pre-formed fibrils in CD163 knockout (KO) mice and wild-type littermates. CD163KO females revealed an impaired and differential early immune response to α-syn pathology as revealed by immunohistochemical and transcriptomic analysis. After 6 months, CD163KO females showed an exacerbated immune response and α-syn pathology, which ultimately led to dopaminergic neurodegeneration of greater magnitude. These findings support a sex-dimorphic neuroprotective role for CD163 during α-syn-induced neurodegeneration.
Collapse
Affiliation(s)
- Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Conghui Li
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ida H Klæstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Zagorka Vitic
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | | | - Asger Kirkegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Gitte U Toft
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Pia Svendsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren K Moestrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Yi S, Wang L, Wang H, Ho MS, Zhang S. Pathogenesis of α-Synuclein in Parkinson's Disease: From a Neuron-Glia Crosstalk Perspective. Int J Mol Sci 2022; 23:14753. [PMID: 36499080 PMCID: PMC9739123 DOI: 10.3390/ijms232314753] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder. The classical behavioral defects of PD patients involve motor symptoms such as bradykinesia, tremor, and rigidity, as well as non-motor symptoms such as anosmia, depression, and cognitive impairment. Pathologically, the progressive loss of dopaminergic (DA) neurons in the substantia nigra (SN) and the accumulation of α-synuclein (α-syn)-composed Lewy bodies (LBs) and Lewy neurites (LNs) are key hallmarks. Glia are more than mere bystanders that simply support neurons, they actively contribute to almost every aspect of neuronal development and function; glial dysregulation has been implicated in a series of neurodegenerative diseases including PD. Importantly, amounting evidence has added glial activation and neuroinflammation as new features of PD onset and progression. Thus, gaining a better understanding of glia, especially neuron-glia crosstalk, will not only provide insight into brain physiology events but also advance our knowledge of PD pathologies. This review addresses the current understanding of α-syn pathogenesis in PD, with a focus on neuron-glia crosstalk. Particularly, the transmission of α-syn between neurons and glia, α-syn-induced glial activation, and feedbacks of glial activation on DA neuron degeneration are thoroughly discussed. In addition, α-syn aggregation, iron deposition, and glial activation in regulating DA neuron ferroptosis in PD are covered. Lastly, we summarize the preclinical and clinical therapies, especially targeting glia, in PD treatments.
Collapse
Affiliation(s)
| | | | | | - Margaret S. Ho
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shiping Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
11
|
García-Revilla J, Herrera AJ, de Pablos RM, Venero JL. Inflammatory Animal Models of Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S165-S182. [PMID: 35662128 PMCID: PMC9535574 DOI: 10.3233/jpd-213138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Accumulating evidence suggests that microglia and peripheral immune cells may play determinant roles in the pathogenesis of Parkinson’s disease (PD). Consequently, there is a need to take advantage of immune-related models of PD to study the potential contribution of microglia and peripheral immune cells to the degeneration of the nigrostriatal system and help develop potential therapies for PD. In this review, we have summarised the main PD immune models. From a historical perspective, we highlight first the main features of intranigral injections of different pro-inflammogens, including lipopolysaccharide (LPS), thrombin, neuromelanin, etc. The use of adenoviral vectors to promote microglia-specific overexpression of different molecules in the ventral mesencephalon, including α-synuclein, IL-1β, and TNF, are also presented and briefly discussed. Finally, we summarise different models associated with peripheral inflammation whose contribution to the pathogenesis of neurodegenerative diseases is now an outstanding question. Illustrative examples included systemic LPS administration and dextran sulfate sodium-induced colitis in rodents.
Collapse
Affiliation(s)
- Juan García-Revilla
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio J. Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rocío M. de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luis Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
12
|
Abstract
Microglia are the primary resident immune cells of the central nervous system. Neuropathological reports have identified augmented microglial activation in brains of patients with neurodegenerative disorders including Parkinson’s disease (PD). Extensive research over the years has strengthened the current view on microglia as a player in the pathogenesis of PD and other α-synucleinopathies. In this review, we summarize key findings of the recent three years on microglia in PD with specific relevance to understanding its heterogeneity, dual nature, and specific interactions with pathological α-synuclein strains to mediate its clearance and spreading. This review provides evidence on the relevance of microglia as a putative biomarker and therapeutic target in PD and related disorders.
Collapse
Affiliation(s)
- Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| |
Collapse
|
13
|
Haverkamp S, Reinhard K, Peichl L, Mietsch M. No evidence for age-related alterations in the marmoset retina. Front Neuroanat 2022; 16:945295. [PMID: 36120100 PMCID: PMC9479465 DOI: 10.3389/fnana.2022.945295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/13/2022] [Indexed: 12/19/2022] Open
Abstract
The physiological aging process of the retina is accompanied by various and sometimes extensive changes: Macular degeneration, retinopathies and glaucoma are the most common findings in the elderly and can potentially lead to irreversible visual disablements up to blindness. To study the aging process and to identify possible therapeutic targets to counteract these diseases, the use of appropriate animal models is mandatory. Besides the most commonly used rodent species, a non-human primate, the common marmoset (Callithrix jacchus) emerged as a promising animal model of human aging over the last years. However, the visual aging process in this species is only partially characterized, especially with regard to retinal aberrations. Therefore, we assessed here for the first time potential changes in retinal morphology of the common marmoset of different age groups. By cell type specific immunolabeling, we analyzed different cell types and distributions, potential photoreceptor and ganglion cell loss, and structural reorganization. We detected no signs of age-related differences in staining patterns or densities of various cell populations. For example, there were no signs of photoreceptor degeneration, and there was only minimal sprouting of rod bipolar cells in aged retinas. Altogether, we describe here the maintenance of a stable neuronal architecture, distribution and number of different cell populations with only mild aberrations during the aging process in the common marmoset retina. These findings are in stark contrast to previously reported findings in rodent species and humans and deserve further investigations to identify the underlying mechanisms and possible therapeutic targets.
Collapse
Affiliation(s)
- Silke Haverkamp
- Department of Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior—Caesar, Bonn, Germany
| | - Katja Reinhard
- Retinal Circuits and Optogenetics, Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Leo Peichl
- Institute of Clinical Neuroanatomy, Dr. Senckenbergische Anatomie, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Matthias Mietsch
- Laboratory Animal Science Unit, German Primate Center, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
Miller SJ, Campbell CE, Jimenez-Corea HA, Wu GH, Logan R. Neuroglial Senescence, α-Synucleinopathy, and the Therapeutic Potential of Senolytics in Parkinson’s Disease. Front Neurosci 2022; 16:824191. [PMID: 35516803 PMCID: PMC9063319 DOI: 10.3389/fnins.2022.824191] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/22/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is the most common movement disorder and the second most prevalent neurodegenerative disease after Alzheimer’s disease. Despite decades of research, there is still no cure for PD and the complicated intricacies of the pathology are still being worked out. Much of the research on PD has focused on neurons, since the disease is characterized by neurodegeneration. However, neuroglia has become recognized as key players in the health and disease of the central nervous system. This review provides a current perspective on the interactive roles that α-synuclein and neuroglial senescence have in PD. The self-amplifying and cyclical nature of oxidative stress, neuroinflammation, α-synucleinopathy, neuroglial senescence, neuroglial chronic activation and neurodegeneration will be discussed. Finally, the compelling role that senolytics could play as a therapeutic avenue for PD is explored and encouraged.
Collapse
Affiliation(s)
- Sean J. Miller
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
| | | | | | - Guan-Hui Wu
- Department of Neurology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Robert Logan
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
- *Correspondence: Robert Logan,
| |
Collapse
|
15
|
Changes in CD163+, CD11b+, and CCR2+ peripheral monocytes relate to Parkinson's disease and cognition. Brain Behav Immun 2022; 101:182-193. [PMID: 35026420 DOI: 10.1016/j.bbi.2022.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein pathology is associated with immune activation and neurodegeneration in Parkinson's disease. The immune activation involves not only microglia but also peripheral immune cells, such as mononuclear phagocytes found in blood and infiltrated in the brain. Understanding peripheral immune involvement is essential for developing immunomodulatory treatment. Therefore, we aimed to study circulating mononuclear phagocytes in early- and late-stage Parkinson's disease, defined by disease duration of less or more than five years, respectively, and analyze their association with clinical phenotypes. We performed a cross-sectional multi-color flow cytometry study on 78 sex-balanced individuals with sporadic Parkinson's disease, 28 controls, and longitudinal samples from seven patients and one control. Cell frequencies and surface marker expressions on natural killer cells, monocyte subtypes, and dendritic cells were compared between groups and correlated with standardized clinical scores. We found elevated frequencies and surface levels of migration- (CCR2, CD11b) and phagocytic- (CD163) markers, particularly on classical and intermediate monocytes in early Parkinson's disease. HLA-DR expression was increased in advanced stages of the disease, whereas TLR4 expression was decreased in women with Parkinson's Disease. The disease-associated immune changes of CCR2 and CD11b correlated with worse cognition. Increased TLR2 expression was related to worse motor symptoms. In conclusion, our data highlights the TLR2 relevance in the symptomatic motor presentation of the disease and a role for peripheral CD163+ and migration-competent monocytes in Parkinson's disease cognitive defects. Our study suggests that the peripheral immune system is dynamically altered in Parkinson's disease stages and directly related to both symptoms and the sex bias of the disease.
Collapse
|
16
|
Bancroft EA, Srinivasan R. Emerging Roles for Aberrant Astrocytic Calcium Signals in Parkinson's Disease. Front Physiol 2022; 12:812212. [PMID: 35087422 PMCID: PMC8787054 DOI: 10.3389/fphys.2021.812212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/09/2021] [Indexed: 11/25/2022] Open
Abstract
Astrocytes display a plethora of spontaneous Ca2+ signals that modulate vital functions of the central nervous system (CNS). This suggests that astrocytic Ca2+ signals also contribute to pathological processes in the CNS. In this context, the molecular mechanisms by which aberrant astrocytic Ca2+ signals trigger dopaminergic neuron loss during Parkinson's disease (PD) are only beginning to emerge. Here, we provide an evidence-based perspective on potential mechanisms by which aberrant astrocytic Ca2+ signals can trigger dysfunction in three distinct compartments of the brain, viz., neurons, microglia, and the blood brain barrier, thereby leading to PD. We envision that the coming decades will unravel novel mechanisms by which aberrant astrocytic Ca2+ signals contribute to PD and other neurodegenerative processes in the CNS.
Collapse
Affiliation(s)
- Eric A. Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, United States
| |
Collapse
|
17
|
Fleming SM, Davis A, Simons E. Targeting alpha-synuclein via the immune system in Parkinson's disease: Current vaccine therapies. Neuropharmacology 2022; 202:108870. [PMID: 34742741 DOI: 10.1016/j.neuropharm.2021.108870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 02/09/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and is defined pathologically by the abnormal accumulation of the presynaptic protein alpha-synuclein (aSyn) in the form of Lewy bodies and Lewy neurites and loss of midbrain dopaminergic neurons in the substantia nigra pars compacta. Because of aSyn's involvement in both sporadic and familial forms of PD, it has become a key target for the development of novel therapeutics. Aberrant aSyn is associated with multiple mechanisms of neuronal dysfunction and degeneration including inflammation, impaired mitochondrial function, altered protein degradation systems, and oxidative stress. Inflammation, in particular, has emerged as a potential significant contributor early in the disease making it an attractive target for disease modification and neuroprotection. Thus, immunotherapies targeting aSyn are currently being investigated in pre-clinical and clinical trials. The focus of this review is to highlight the role of aSyn in neuroinflammation and discuss the current status of aSyn-directed immunotherapies in pre-clinical and clinical trials for PD.
Collapse
Affiliation(s)
- Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, USA.
| | - Ashley Davis
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, USA
| | - Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, USA
| |
Collapse
|
18
|
α-Synuclein Overexpression Increases Dopamine D2/3 Receptor Binding and Immune Activation in a Model of Early Parkinson’s Disease. Biomedicines 2021; 9:biomedicines9121876. [PMID: 34944691 PMCID: PMC8698691 DOI: 10.3390/biomedicines9121876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Progressive degeneration of dopaminergic neurons, immune activation, and α-synuclein pathology characterize Parkinson’s disease (PD). We previously reported that unilateral intranigral injection of recombinant adeno-associated viral (rAAV) vectors encoding wild-type human α-synuclein produced a rat model of early PD with dopamine terminal dysfunction. Here we tested the hypothesis that decreases in dopamine result in increased postsynaptic dopamine D2/D3 receptor expression, neuroinflammation, and reduced synaptic vesicle glycoprotein 2A (SV2A) density. Rats were injected with rAAV encoding α-synuclein or green fluorescent protein and subjected to non-pharmacological motor tests, before euthanization at 12 weeks post-injection. We performed: (1) in situ hybridization of nigral tyrosine hydroxylase mRNA, (2) HPLC of striatal dopamine content, and (3) autoradiography with [3H]raclopride, [3H]DTBZ, [3H]GBR12935, [3H]PK11195, and [3H]UCB-J to measure binding at D2/3 receptors, vesicular monoamine transporter 2, dopamine transporters, mitochondrial translocator protein, and SV2A, respectively. rAAV-α-synuclein induced motor asymmetry and reduced tyrosine hydroxylase mRNA and dopamine content in ipsilateral brain regions. This was paralleled by elevated ipsilateral postsynaptic dopamine D2/3 receptor expression and immune activation, with no changes to synaptic SV2A density. In conclusion, α-synuclein overexpression results in dopaminergic degeneration that induced compensatory increases in D2/3 binding and immune activation, recapitulating many of the pathological characteristics of PD.
Collapse
|
19
|
Neuroinflammation in Parkinson's disease: a meta-analysis of PET imaging studies. J Neurol 2021; 269:2304-2314. [PMID: 34724571 DOI: 10.1007/s00415-021-10877-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
Abstract
Increasingly, evidence implicates an important role of neuroinflammation in neurodegeneration progression. Yet, brain imaging has not reached a consistent conclusion that neuroinflammation is involved in the pathogenesis of Parkinson's disease (PD). We aimed to review the evidence to quantitatively assess the existence and spatial distribution of neuroinflammation in the brain of PD patients. We systematically searched literature databases for case-control studies which used positron emission tomography to detect neuroinflammation represented by translocator protein (TSPO) levels in PD patients compared with healthy controls (HC). Standardized mean differences (SMD) were selected as effect sizes and random-effects models were used to combine effect sizes. Subgroup analyses for separate brain regions were conducted. Fifteen studies comprising 455 (HC = 198, PD = 238) participants and 19 brain regions were included. Compared to HC, PD patients had elevated TSPO levels in midbrain, putamen, anterior cingulate, posterior cingulate, thalamus, striatum, frontal, temporal, parietal, occipital, cortex, hippocampus, substantia nigra, pons, cerebellum, and caudate when using 1st-generation ligands. TSPO levels were elevated in the midbrain of PD patients when 2nd-generation ligands were used. We discussed the possible explanations of contrasting difference between these outcomes.
Collapse
|
20
|
Selective targeting of the TLR2/MyD88/NF-κB pathway reduces α-synuclein spreading in vitro and in vivo. Nat Commun 2021; 12:5382. [PMID: 34508096 PMCID: PMC8433339 DOI: 10.1038/s41467-021-25767-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/31/2021] [Indexed: 02/07/2023] Open
Abstract
Pathways to control the spreading of α-synuclein (α-syn) and associated neuropathology in Parkinson’s disease (PD), multiple system atrophy (MSA) and dementia with Lewy bodies (DLB) are unclear. Here, we show that preformed α-syn fibrils (PFF) increase the association between TLR2 and MyD88, resulting in microglial activation. The TLR2-interaction domain of MyD88 (wtTIDM) peptide-mediated selective inhibition of TLR2 reduces PFF-induced microglial inflammation in vitro. In PFF-seeded A53T mice, the nasal administration of the wtTIDM peptide, NEMO-binding domain (wtNBD) peptide, or genetic deletion of TLR2 reduces glial inflammation, decreases α-syn spreading, and protects dopaminergic neurons by inhibiting NF-κB. In summary, α-syn spreading depends on the TLR2/MyD88/NF-κB pathway and it can be reduced by nasal delivery of wtTIDM and wtNBD peptides. The mechanisms underlying the spreading of α-synuclein in various α-synucleinopathies are unclear. Here, the authors show that targeting the TLR2/MyD88/NF-κB pathway can reduce α-synuclein spreading, reduce neuroinflammation, and protect dopaminergic neurons in vitro and in mouse models
Collapse
|
21
|
Gentzel RC, Toolan D, Jinn S, Schachter JB, Ma L, Kahle PJ, Smith SM, Marcus JN. Intracranial administration of alpha-synuclein fibrils in A30P-synuclein transgenic mice causes robust synucleinopathy and microglial induction. Neurobiol Aging 2021; 106:12-25. [PMID: 34225000 DOI: 10.1016/j.neurobiolaging.2021.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/12/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022]
Abstract
Synucleinopathies are neurodegenerative disorders involving pathological alpha-synuclein (αSyn) protein, including dementia with Lewy bodies, multiple system atrophy and Parkinson's disease (PD). Current in vivo models of synucleinopathy include transgenic mice overexpressing αSyn variants and methods based on administration of aggregated, exogenous αSyn. Combining these techniques offers the ability to study consequences of introducing pathological αSyn into primed neuronal environments likely to develop synucleinopathy. Herein, we characterize the impacts pre-formed fibrils (PFFs) of recombinant, human αSyn have in mice overexpressing human A30P αSyn, a mutation associated with autosomal dominant PD. A30P mouse brain contains detergent insoluble αSyn biochemically similar to PD brain, and these mice develop Lewy-like synucleinopathy with age. Administration of PFFs in A30P mice resulted in regionally-specific accumulations of phosphorylated synuclein, microglial induction and a motor phenotype that differed from PFF-induced effects in wildtype mice. Surprisingly, PFF-induced losses of tyrosine hydroxylase were similar in A30P and wildtype mice. Thus, the PFF-A30P model recapitulates key aspects of synucleinopathy with induction of microglia, creating an appropriate system for evaluating neurodegenerative therapeutics.
Collapse
Affiliation(s)
- Renee C Gentzel
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Dawn Toolan
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sarah Jinn
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Joel B Schachter
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA; Currently at Takeda Pharmaceutics, Inc., San Diego, CA, USA
| | - Lei Ma
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Philipp J Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research and, German Center for Neurodegenerative Diseases, University of Tübingen, Germany
| | - Sean M Smith
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jacob N Marcus
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
22
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
23
|
Preclinical Marmoset Model for Targeting Chronic Inflammation as a Strategy to Prevent Alzheimer's Disease. Vaccines (Basel) 2021; 9:vaccines9040388. [PMID: 33920929 PMCID: PMC8071309 DOI: 10.3390/vaccines9040388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 11/17/2022] Open
Abstract
Due to the aging population, modern society is facing an increasing prevalence of neurological diseases such as Alzheimer’s disease (AD). AD is an age-related chronic neurodegenerative disorder for which no satisfying therapy exists. Understanding the mechanisms underlying the onset of AD is necessary to find targets for protective treatment. There is growing awareness of the essential role of the immune system in the early AD pathology. Amyloidopathy, the main feature of early-stage AD, has a deregulating effect on the immune function. This is reciprocal as the immune system also affects amyloidopathy. It seems that the inflammatory reaction shows a heterogeneous pattern depending on the stage of the disease and the variation between individuals, making not only the target but also the timing of treatment important. The lack of relevant translational animal models that faithfully reproduce clinical and pathogenic features of AD is a major cause of the delay in developing new disease-modifying therapies and their optimal timing of administration. This review describes the communication between amyloidopathy and inflammation and the possibility of using nonhuman primates as a relevant animal model for preclinical AD research.
Collapse
|
24
|
Harms AS, Ferreira SA, Romero-Ramos M. Periphery and brain, innate and adaptive immunity in Parkinson's disease. Acta Neuropathol 2021; 141:527-545. [PMID: 33555429 PMCID: PMC7952334 DOI: 10.1007/s00401-021-02268-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 12/29/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where alpha-synuclein plays a central role in the death and dysfunction of neurons, both, in central, as well as in the peripheral nervous system. Besides the neuronal events observed in patients, PD also includes a significant immune component. It is suggested that the PD-associated immune response will have consequences on neuronal health, thus opening immunomodulation as a potential therapeutic strategy in PD. The immune changes during the disease occur in the brain, involving microglia, but also in the periphery with changes in cells of the innate immune system, particularly monocytes, as well as those of adaptive immunity, such as T-cells. This realization arises from multiple patient studies, but also from data in animal models of the disease, providing strong evidence for innate and adaptive immune system crosstalk in the central nervous system and periphery in PD. Here we review the data showing that alpha-synuclein plays a crucial role in the activation of the innate and adaptive immune system. We will also describe the studies suggesting that inflammation in PD includes early changes in innate and adaptive immune cells that develop dynamically through time during disease, contributing to neuronal degeneration and symptomatology in patients. This novel finding has contributed to the definition of PD as a multisystem disease that should be approached in a more integratory manner rather than a brain-focused classical approach.
Collapse
Affiliation(s)
- Ashley S Harms
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara A Ferreira
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark.
| |
Collapse
|
25
|
Kam TI, Hinkle JT, Dawson TM, Dawson VL. Microglia and astrocyte dysfunction in parkinson's disease. Neurobiol Dis 2020; 144:105028. [PMID: 32736085 PMCID: PMC7484088 DOI: 10.1016/j.nbd.2020.105028] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 07/01/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
While glia are essential for regulating the homeostasis in the normal brain, their dysfunction contributes to neurodegeneration in many brain diseases, including Parkinson's disease (PD). Recent studies have identified that PD-associated genes are expressed in glial cells as well as neurons and have crucial roles in microglia and astrocytes. Here, we discuss the role of microglia and astrocytes dysfunction in relation to PD-linked mutations and their implications in PD pathogenesis. A better understanding of microglia and astrocyte functions in PD may provide insights into neurodegeneration and novel therapeutic approaches for PD.
Collapse
Affiliation(s)
- Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jared T Hinkle
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Fouka M, Mavroeidi P, Tsaka G, Xilouri M. In Search of Effective Treatments Targeting α-Synuclein Toxicity in Synucleinopathies: Pros and Cons. Front Cell Dev Biol 2020; 8:559791. [PMID: 33015057 PMCID: PMC7500083 DOI: 10.3389/fcell.2020.559791] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB) represent pathologically similar, progressive neurodegenerative disorders characterized by the pathological aggregation of the neuronal protein α-synuclein. PD and DLB are characterized by the abnormal accumulation and aggregation of α-synuclein in proteinaceous inclusions within neurons named Lewy bodies (LBs) and Lewy neurites (LNs), whereas in MSA α-synuclein inclusions are mainly detected within oligodendrocytes named glial cytoplasmic inclusions (GCIs). The presence of pathologically aggregated α-synuclein along with components of the protein degradation machinery, such as ubiquitin and p62, in LBs and GCIs is considered to underlie the pathogenic cascade that eventually leads to the severe neurodegeneration and neuroinflammation that characterizes these diseases. Importantly, α-synuclein is proposed to undergo pathogenic misfolding and oligomerization into higher-order structures, revealing self-templating conformations, and to exert the ability of "prion-like" spreading between cells. Therefore, the manner in which the protein is produced, is modified within neural cells and is degraded, represents a major focus of current research efforts in the field. Given that α-synuclein protein load is critical to disease pathogenesis, the identification of means to limit intracellular protein burden and halt α-synuclein propagation represents an obvious therapeutic approach in synucleinopathies. However, up to date the development of effective therapeutic strategies to prevent degeneration in synucleinopathies is limited, due to the lack of knowledge regarding the precise mechanisms underlying the observed pathology. This review critically summarizes the recent developed strategies to counteract α-synuclein toxicity, including those aimed to increase protein degradation, to prevent protein aggregation and cell-to-cell propagation, or to engage antibodies against α-synuclein and discuss open questions and unknowns for future therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
27
|
Wang L, Liu Y, Yan S, Du T, Fu X, Gong X, Zhou X, Zhang T, Wang X. Disease Progression-Dependent Expression of CD200R1 and CX3CR1 in Mouse Models of Parkinson's Disease. Aging Dis 2020; 11:254-268. [PMID: 32257540 PMCID: PMC7069458 DOI: 10.14336/ad.2019.0615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022] Open
Abstract
Microglial activation is an important contributor to the pathogenesis of Parkinson’s disease (PD). Microglia are tightly and efficiently regulated by immune checkpoints, including CD200-CD200R1 and CX3CL1-CX3CR1. Understanding the involvement of these checkpoints in disease progression provides important insights into how microglial activation contributes to PD pathology. However, so far, studies have produced seemingly conflicting results. In this study, we demonstrate that CD200R1 expression is down-regulated at both early and late stage of PD model, and CX3CR1 expression is down-regulated in early stage and recovered in late stage. In primary cultured microglia, CD200R1 and CX3CR1 expressions are both directly regulated by LPS or α-synuclein, and CD200R1 expression is more sensitively regulated than CX3CR1. In addition, CD200 knockout causes an increase in proinflammatory cytokine production and microglial activation in the midbrain. Remarkably, DA neurons in the substantial nigra are degenerated in CD200-/- mice. Finally, activation of the CD200R with CD200Fc alleviates the neuroinflammation in microglia. Together, these results suggest that immune checkpoints play distinct functional roles in different stage of PD pathology, and the CD200-CD200R1 axis plays a significant role in nigrostriatal neuron viability and function.
Collapse
Affiliation(s)
- Le Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yang Liu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Shuxin Yan
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xia Fu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- 2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Xinyu Zhou
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Role of brain renin angiotensin system in neurodegeneration: An update. Saudi J Biol Sci 2020; 27:905-912. [PMID: 32127770 PMCID: PMC7042626 DOI: 10.1016/j.sjbs.2020.01.026] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 01/12/2023] Open
Abstract
Renin angiotensin system (RAS) is an endocrine system widely known for its physiological roles in electrolyte homeostasis, body fluid volume regulation and cardiovascular control in peripheral circulation. However, brain RAS is an independent form of RAS expressed locally in the brain, which is known to be involved in brain functions and disorders. There is strong evidence for a major involvement of excessive brain angiotensin converting enzyme (ACE)/Angiotensin II (Ang II)/Angiotensin type-1 receptor (AT-1R) axis in increased activation of oxidative stress, apoptosis and neuroinflammation causing neurodegeneration in several brain disorders. Numerous studies have demonstrated strong neuroprotective effects by blocking AT1R in these brain disorders. Additionally, the angiotensin converting enzyme 2 (ACE2)/Angiotensin (1–7)/Mas receptor (MASR), is another axis of brain RAS which counteracts the damaging effects of ACE/Ang II/AT1R axis on neurons in the brain. Thus, angiotensin II receptor blockers (ARBs) and activation of ACE2/Angiotensin (1–7)/MASR axis may serve as an exciting and novel method for neuroprotection in several neurodegenerative diseases. Here in this review article, we discuss the expression of RAS in the brain and highlight how altered RAS level may cause neurodegeneration. Understanding the pathophysiology of RAS and their links to neurodegeneration has enormous potential to identify potentially effective pharmacological tools to treat neurodegenerative diseases in the brain.
Collapse
|
29
|
Olsen LK, Cairns AG, Ådén J, Moriarty N, Cabre S, Alamilla VR, Almqvist F, Dowd E, McKernan DP. Viral mimetic priming enhances α-synuclein-induced degeneration: Implications for Parkinson's disease. Brain Behav Immun 2019; 80:525-535. [PMID: 31029796 DOI: 10.1016/j.bbi.2019.04.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/29/2019] [Accepted: 04/24/2019] [Indexed: 11/19/2022] Open
Abstract
Evidence is accumulating to suggest that viral infections and consequent viral-mediated neuroinflammation may contribute to the etiology of idiopathic Parkinson's disease. Moreover, viruses have been shown to influence α-synuclein oligomerization as well as the autophagic clearance of abnormal intra-cellular proteins aggregations, both of which are key neuropathological events in Parkinson's disease pathogenesis. To further investigate the interaction between viral-mediated neuroinflammation and α-synuclein aggregation in the context of Parkinson's disease, this study sought to determine the impact of viral neuroinflammatory priming on α-synuclein aggregate-induced neuroinflammation and neurotoxicity in the rat nigrostriatal pathway. To do so, male Sprague-Dawley rats were intra-nigrally injected with a synthetic mimetic of viral dsRNA (poly I:C) followed two weeks later by a peptidomimetic small molecule which accelerates α-synuclein fibril formation (FN075). The impact of the viral priming on α-synuclein aggregation-induced neuroinflammation, neurodegeneration and motor dysfunction was assessed. We found that prior administration of the viral mimetic poly I:C significantly exacerbated or precipitated the α-synuclein aggregate induced neuropathological and behavioral effects. Specifically, sequential exposure to the two challenges caused a significant increase in nigral microgliosis (p < 0.001) and astrocytosis (p < 0.01); precipitated a significant degeneration of the nigrostriatal cell bodies (p < 0.05); and precipitated a significant impairment in forelimb kinesis (p < 0.01) and sensorimotor integration (p < 0.01). The enhanced sensitivity of the nigrostriatal neurons to pathological α-synuclein aggregation after viral neuroinflammatory priming further suggests that viral infections may contribute to the etiology and pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Laura K Olsen
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland
| | | | - Jörgen Ådén
- Department of Chemistry, Umeå University, Sweden
| | - Niamh Moriarty
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland
| | - Silvia Cabre
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland
| | - Veronica R Alamilla
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland
| | | | - Eilís Dowd
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland
| | - Declan P McKernan
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, Ireland.
| |
Collapse
|
30
|
Refolo V, Stefanova N. Neuroinflammation and Glial Phenotypic Changes in Alpha-Synucleinopathies. Front Cell Neurosci 2019; 13:263. [PMID: 31263402 PMCID: PMC6585624 DOI: 10.3389/fncel.2019.00263] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023] Open
Abstract
The role of neuroinflammation has been increasingly recognized in the field of neurodegenerative diseases. Many studies focusing on the glial cells involved in the inflammatory responses of the brain, namely microglia and astroglia, have over the years pointed out the dynamic and changing behavior of these cells, accompanied by different morphologies and activation forms. This is particularly evident in diseased conditions, where glia react to any shift from homeostasis, acquiring different phenotypes. Particularly for microglia, it has soon become clear that such phenotypes are multiple, as multiple are the functions related to them. Several approaches have over time revealed different facets of microglial phenotypic diversity, and advanced genetic analyses, in recent years, have added new insights into microglial heterogeneity, opening novel scenarios that researchers have just started to explore. Among neurodegenerative diseases, an important section is represented by alpha-synucleinopathies. Here alpha-synuclein accumulates abnormally in the brain and, depending on its pattern of distribution, leads to the development of different clinical conditions. Also for these proteinopathies, neuroinflammation and glial activation have been identified as constant and crucial factors during disease development. In the present review we will address the current literature about glial phenotypic changes with respect to alpha-synucleinopathies, as well as consider the pathophysiological and therapeutic implications of such a dynamic cellular behavior.
Collapse
Affiliation(s)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Fuzzati-Armentero MT, Cerri S, Blandini F. Peripheral-Central Neuroimmune Crosstalk in Parkinson's Disease: What Do Patients and Animal Models Tell Us? Front Neurol 2019; 10:232. [PMID: 30941089 PMCID: PMC6433876 DOI: 10.3389/fneur.2019.00232] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is no longer considered an immune privileged organ and neuroinflammation has long been associated with Parkinson's disease. Accumulating evidence demonstrates that innate and adaptive responses take place in the CNS. The extent to which peripheral immune alterations impacts on the CNS, or vice and versa, is, however, still a matter of debate. Gaining a better knowledge of the molecular and cellular immune dysfunctions present in these two compartments and clarifying their mutual interactions is a fundamental step in understanding and preventing Parkinson's disease (PD) pathogenesis. This review provides an overview of the current knowledge on inflammatory processes evidenced both in PD patients and in toxin-induced animal models of the disease. It discusses differences and similarities between human and animal studies in the context of neuroinflammation and immune responses and how they have guided therapeutic strategies to slow down disease progression. Future longitudinal studies are necessary and can help gain a better understanding on peripheral-central nervous system crosstalk to improve therapeutic strategies for PD.
Collapse
|
32
|
Tansey MG, Romero-Ramos M. Immune system responses in Parkinson's disease: Early and dynamic. Eur J Neurosci 2019; 49:364-383. [PMID: 30474172 PMCID: PMC6391192 DOI: 10.1111/ejn.14290] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
Abstract
The neuropathological hallmarks of Parkinson's disease (PD) are the degeneration and death of dopamine-producing neurons in the ventral midbrain, the widespread intraneuronal aggregation of alpha-synuclein (α) in Lewy bodies and neurites, neuroinflammation, and gliosis. Signs of microglia activation in the PD brain postmortem as well as during disease development revealed by neuroimaging, implicate immune responses in the pathophysiology of the disease. Intensive research during the last two decades has advanced our understanding of the role of these responses in the disease process, yet many questions remain unanswered. A transformative finding in the field has been the confirmation that in vivo microglia are able to respond directly to pathological a-syn aggregates but also to neuronal dysfunction due to intraneuronal a-syn toxicity well in advance of neuronal death. In addition, clinical research and disease models have revealed the involvement of both the innate and adaptive immune systems. Indeed, the data suggest that PD leads not only to a microglia response, but also to a cellular and humoral peripheral immune response. Together, these findings compel us to consider a more holistic view of the immunological processes associated with the disease. Central and peripheral immune responses aimed at maintaining neuronal health will ultimately have consequences on neuronal survival. We will review here the most significant findings that have contributed to the current understanding of the immune response in PD, which is proposed to occur early, involve peripheral and brain immune cells, evolve as neuronal dysfunction progresses, and is likely to influence disease progression.
Collapse
Affiliation(s)
- Malú G Tansey
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Marina Romero-Ramos
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, & AU IDEAS center NEURODIN, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
33
|
Rodriguez-Perez AI, Sucunza D, Pedrosa MA, Garrido-Gil P, Kulisevsky J, Lanciego JL, Labandeira-Garcia JL. Angiotensin Type 1 Receptor Antagonists Protect Against Alpha-Synuclein-Induced Neuroinflammation and Dopaminergic Neuron Death. Neurotherapeutics 2018; 15:1063-1081. [PMID: 29987762 PMCID: PMC6277291 DOI: 10.1007/s13311-018-0646-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The loss of dopaminergic neurons and α-synuclein accumulation are major hallmarks of Parkinson's disease (PD), and it has been suggested that a major mechanism of α-synuclein toxicity is microglial activation. The lack of animal models that properly reproduce PD, and particularly the underlying synucleinopathy, has hampered the clarification of PD mechanisms and the development of effective therapies. Here, we used neurospecific adeno-associated viral vectors serotype 9 coding for either the wild-type or mutated forms of human alpha-synuclein (WT and SynA53T, respectively) under the control of a synapsin promoter to further induce a marked dopaminergic neuron loss together with an important microglial neuroinflammatory response. Overexpression of neuronal alpha-synuclein led to increased expression of angiotensin type 1 receptors and NADPH oxidase activity, together with a marked increase in the number of OX-6-positive microglial cells and expression of markers of phagocytic activity (CD68) and classical pro-inflammatory/M1 microglial phenotype markers such as inducible nitric oxide synthase, tumor necrosis factor alpha, interleukin-1β, and IL-6. Moreover, a significant decrease in the expression of markers of immunoregulatory/M2 microglial phenotype such as the enzyme arginase-1 was constantly observed. Interestingly, alpha-synuclein-induced changes in microglial phenotype markers and dopaminergic neuron death were inhibited by simultaneous treatment with the angiotensin type 1 blockers candesartan or telmisartan. Our results suggest the repurposing of candesartan and telmisartan as a neuroprotective strategy for PD.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
| | - Diego Sucunza
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
- Neurosciences Division, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, 31008, Spain
| | - Maria A Pedrosa
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
| | - Jaime Kulisevsky
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital and Biomedical Research Institute, Universitat Autonoma de Barcelona and Universitat Oberta de Catalunya, Barcelona, 08025, Spain
| | - Jose L Lanciego
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain
- Neurosciences Division, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, 31008, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas), Madrid, 28031, Spain.
| |
Collapse
|
34
|
Lillethorup TP, Glud AN, Landeck N, Alstrup AKO, Jakobsen S, Vang K, Doudet DJ, Brooks DJ, Kirik D, Hinz R, Sørensen JC, Landau AM. In vivo quantification of glial activation in minipigs overexpressing human α-synuclein. Synapse 2018; 72:e22060. [PMID: 30009467 DOI: 10.1002/syn.22060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/06/2018] [Accepted: 07/11/2018] [Indexed: 12/25/2022]
Abstract
Parkinson's disease is characterized by a progressive loss of substantia nigra (SN) dopaminergic neurons and the formation of Lewy bodies containing accumulated alpha-synuclein (α-syn). The pathology of Parkinson's disease is associated with neuroinflammatory microglial activation, which may contribute to the ongoing neurodegeneration. This study investigates the in vivo microglial and dopaminergic response to overexpression of α-syn. We used positron emission tomography (PET) and the 18 kDa translocator protein radioligand, [11 C](R)PK11195, to image brain microglial activation and (+)-α-[11 C]dihydrotetrabenazine ([11 C]DTBZ), to measure vesicular monoamine transporter 2 (VMAT2) availability in Göttingen minipigs following injection with recombinant adeno-associated virus (rAAV) vectors expressing either mutant A53T α-syn or green fluorescent protein (GFP) into the SN (4 rAAV-α-syn, 4 rAAV-GFP, 5 non-injected control minipigs). We performed motor symptom assessment and immunohistochemical examination of tyrosine hydroxylase (TH) and transgene expression. Expression of GFP and α-syn was observed at the SN injection site and in the striatum. We observed no motor symptoms or changes in striatal [11 C]DTBZ binding potential in vivo or striatal or SN TH staining in vitro between the groups. The mean [11 C](R)PK11195 total volume of distribution was significantly higher in the basal ganglia and cortical areas of the α-syn group than the control animals. We conclude that mutant α-syn expression in the SN resulted in microglial activation in multiple sub- and cortical regions, while it did not affect TH stains or VMAT2 availability. Our data suggest that microglial activation constitutes an early response to accumulation of α-syn in the absence of dopamine neuron degeneration.
Collapse
Affiliation(s)
- Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Andreas Nørgaard Glud
- Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Natalie Landeck
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Kim Vang
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Doris J Doudet
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Department of Medicine/Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J Brooks
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Division of Neuroscience, Department of Medicine, Imperial College London, London, United Kingdom.,Division of Neuroscience, Newcastle University, Newcastle, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Jens Christian Sørensen
- Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark.,Translational Neuropsychiatry Unit, Institute of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|
35
|
Ferreira SA, Romero-Ramos M. Microglia Response During Parkinson's Disease: Alpha-Synuclein Intervention. Front Cell Neurosci 2018; 12:247. [PMID: 30127724 PMCID: PMC6087878 DOI: 10.3389/fncel.2018.00247] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
The discovery of the central role played by the protein alpha-synuclein in Parkinson's disease and other Lewy body brain disorders has had a great relevance in the understanding of the degenerative process occurring in these diseases. In addition, during the last two decades, the evidence suggesting an immune response in Parkinson's disease patients have multiplied. The role of the immune system in the disease is supported by data from genetic studies and patients, as well as from laboratory animal models and cell cultures. In the immune response, the microglia, the immune cell of the brain, will have a determinant role. Interestingly, alpha-synuclein is suggested to have a central function not only in the neuronal events occurring in Parkinson's disease, but also in the immune response during the disease. Numerous studies have shown that alpha-synuclein can act directly on immune cells, such as microglia in brain, initiating a sterile response that will have consequences for the neuronal health and that could also translate in a peripheral immune response. In parallel, microglia should also act clearing alpha-synuclein thus avoiding an overabundance of the protein, which is crucial to the disease progression. Therefore, the microglia response in each moment will have significant consequences for the neuronal fate. Here we will review the literature addressing the microglia response in Parkinson's disease with an especial focus on the protein alpha-synuclein. We will also reflect upon the limitations of the studies carried so far and in the therapeutic possibilities opened based on these recent findings.
Collapse
Affiliation(s)
- Sara A Ferreira
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
36
|
NCX1 and NCX3 as potential factors contributing to neurodegeneration and neuroinflammation in the A53T transgenic mouse model of Parkinson's Disease. Cell Death Dis 2018; 9:725. [PMID: 29941946 PMCID: PMC6018508 DOI: 10.1038/s41419-018-0775-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022]
Abstract
Na+-Ca2+ exchanger (NCX) isoforms constitute the major cellular Ca2+ extruding system in neurons and microglia. We herein investigated the role of NCX isoforms in the pathophysiology of Parkinson's disease (PD). Their expression and activity were evaluated in neurons and glia of mice expressing the human A53T variant of α-synuclein (A53T mice), an animal model mimicking a familial form of PD. Western blotting revealed that NCX3 expression in the midbrain of 12-month old A53T mice was lower than that of wild type (WT). Conversely, NCX1 expression increased in the striatum. Immunohistochemical studies showed that glial fibrillary acidic protein (GFAP)-positive astroglial cells significantly increased in the substantia nigra pars compacta (SNc) and in the striatum. However, the number and the density of tyrosine hydroxylase (TH)-positive neurons decreased in both brain regions. Interestingly, ionized calcium binding adaptor molecule 1 (IBA-1)-positive microglial cells increased only in the striatum of A53T mice compared to WT. Double immunostaining studies showed that in A53T mice, NCX1 was exclusively co-expressed in IBA-1-positive microglial cells in the striatum, whereas NCX3 was solely co-expressed in TH-positive neurons in SNc. Beam walking and pole tests revealed a reduction in motor performance for A53T mice compared to WT. In vitro experiments in midbrain neurons from A53T and WT mice demonstrated a reduction in NCX3 expression, which was accompanied by mitochondrial overload of Ca2+ ions, monitored with confocal microscopy by X-Rhod-1 fluorescent dye. Collectively, in vivo and in vitro findings suggest that the reduction in NCX3 expression and activity in A53T neurons from midbrain may cause mitochondrial dysfunction and neuronal death in this brain area, whereas NCX1 overexpression in microglial cells may promote their proliferation in the striatum.
Collapse
|
37
|
Henrich MT, Geibl FF, Lee B, Chiu WH, Koprich JB, Brotchie JM, Timmermann L, Decher N, Matschke LA, Oertel WH. A53T-α-synuclein overexpression in murine locus coeruleus induces Parkinson's disease-like pathology in neurons and glia. Acta Neuropathol Commun 2018; 6:39. [PMID: 29747690 PMCID: PMC5946574 DOI: 10.1186/s40478-018-0541-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/25/2018] [Indexed: 12/24/2022] Open
Abstract
Degeneration of noradrenergic locus coeruleus neurons occurs during the prodromal phase of Parkinson's disease and contributes to a variety of non-motor symptoms, e.g. depression, anxiety and REM sleep behavior disorder. This study was designed to establish the first locus coeruleus α-synucleinopathy mouse model, which should provide sufficient information about the time-course of noradrenergic neurodegeneration, replicate cardinal histopathological features of the human Parkinson's disease neuropathology and finally lead to robust histological markers, which are sufficient to assess the pathological changes in a quantitative and qualitative way. We show that targeted viral vector-mediated overexpression of human mutant A53T-α-synuclein in vivo in locus coeruleus neurons of wild-type mice resulted in progressive noradrenergic neurodegeneration over a time frame of 9 weeks. Observed neuronal cell loss was accompanied by progressive α-synuclein phosphorylation, formation of proteinase K-resistant α-synuclein-aggregates, accumulation of Ubi-1- and p62-positive inclusions in microglia and induction of progressive micro- and astrogliosis. Apart from this local pathology, abundant α-synuclein-positive axons were found in locus coeruleus output regions, indicating rapid anterograde axonal transport of A53T-α-synuclein. Taken together, we present the first model of α-synucleinopathy in the murine locus coeruleus, replicating essential morphological features of human Parkinson's disease pathology. This new model may contribute to the research on prodromal Parkinson's disease, in respect to pathophysiology and the development of disease-modifying therapy.
Collapse
|
38
|
Benskey MJ, Sellnow RC, Sandoval IM, Sortwell CE, Lipton JW, Manfredsson FP. Silencing Alpha Synuclein in Mature Nigral Neurons Results in Rapid Neuroinflammation and Subsequent Toxicity. Front Mol Neurosci 2018; 11:36. [PMID: 29497361 PMCID: PMC5819572 DOI: 10.3389/fnmol.2018.00036] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/26/2018] [Indexed: 12/19/2022] Open
Abstract
Human studies and preclinical models of Parkinson’s disease implicate the involvement of both the innate and adaptive immune systems in disease progression. Further, pro-inflammatory markers are highly enriched near neurons containing pathological forms of alpha synuclein (α-syn), and α-syn overexpression recapitulates neuroinflammatory changes in models of Parkinson’s disease. These data suggest that α-syn may initiate a pathological inflammatory response, however the mechanism by which α-syn initiates neuroinflammation is poorly understood. Silencing endogenous α-syn results in a similar pattern of nigral degeneration observed following α-syn overexpression. Here we aimed to test the hypothesis that loss of α-syn function within nigrostriatal neurons results in neuronal dysfunction, which subsequently stimulates neuroinflammation. Adeno-associated virus (AAV) expressing an short hairpin RNA (shRNA) targeting endogenous α-syn was unilaterally injected into the substantia nigra pars compacta (SNc) of adult rats, after which nigrostriatal pathology and indices of neuroinflammation were examined at 7, 10, 14 and 21 days post-surgery. Removing endogenous α-syn from nigrostriatal neurons resulted in a rapid up-regulation of the major histocompatibility complex class 1 (MHC-1) within transduced nigral neurons. Nigral MHC-1 expression occurred prior to any overt cell death and coincided with the recruitment of reactive microglia and T-cells to affected neurons. Following the induction of neuroinflammation, α-syn knockdown resulted in a 50% loss of nigrostriatal neurons in the SNc and a corresponding loss of nigrostriatal terminals and dopamine (DA) concentrations within the striatum. Expression of a control shRNA did not elicit any pathological changes. Silencing α-syn within glutamatergic neurons of the cerebellum did not elicit inflammation or cell death, suggesting that toxicity initiated by α-syn silencing is specific to DA neurons. These data provide evidence that loss of α-syn function within nigrostriatal neurons initiates a neuronal-mediated neuroinflammatory cascade, involving both the innate and adaptive immune systems, which ultimately results in the death of affected neurons.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Rhyomi C Sellnow
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Ivette M Sandoval
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Jack W Lipton
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
39
|
Olesen MN, Christiansen JR, Petersen SV, Jensen PH, Paslawski W, Romero-Ramos M, Sanchez-Guajardo V. CD4 T cells react to local increase of α-synuclein in a pathology-associated variant-dependent manner and modify brain microglia in absence of brain pathology. Heliyon 2018; 4:e00513. [PMID: 29560431 PMCID: PMC5857520 DOI: 10.1016/j.heliyon.2018.e00513] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/03/2018] [Accepted: 01/11/2018] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that immunological processes in the brain during α-synuclein-induced neurodegeneration vary depending on the presence or absence of cell death. This suggests that the immune system is able to react differently to the different stages of α-synuclein pathology. However, it was unclear whether these immune changes were governed by brain processes or by a direct immune response to α-synuclein modifications. We have herein locally increased the peripheral concentration of α-synuclein or its pathology-associated variants, nitrated or fibrillar, to characterize the modulation of the CD4 T cell pool by α-synuclein and brain microglia in the absence of any α-synuclein brain pathology. We observed that α-synuclein changed the CD4:CD8 ratio by contracting the CD3+CD4+ T cell pool and reducing the pool of memory Regulatory T cells (Treg). Nitrated α-synuclein induced the expansion of both the CD3+CD4+ and CD3+CD4- T cells, while fibrils increased the percentage of Foxp3+ Treg cells and induced anti-α-synuclein antibodies. Furthermore, the activation pattern of CD3+CD4+ T cells was modulated in a variant-dependent manner; while nitrated and fibrillar α-synuclein expanded the fraction of activated Treg, all three α-synuclein variants reduced the expression levels of STAT3, CD25 and CD127 on CD3+CD4+ T cells. Additionally, while monomeric α-synuclein increased CD103 expression, the fibrils decreased it, and CCR6 expression was decreased by nitrated and fibrillar α-synuclein, indicating that α-synuclein variants affect the homing and tolerance capacities of CD3+CD4+ T cells. Indeed, this correlated with changes in brain microglia phenotype, as determined by FACS analysis, in an α-synuclein variant-specific manner and coincided in time with CD4+ T cell infiltration into brain parenchyma. We have shown that the peripheral immune system is able to sense and react specifically to changes in the local concentration and structure of α-synuclein, which results in variant-specific T cell migration into the brain. This may have a specific repercussion for brain microglia.
Collapse
Affiliation(s)
- Mads N Olesen
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Josefine R Christiansen
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Steen Vang Petersen
- Laboratory for Redox Regulation, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Wojciech Paslawski
- iNANO, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
40
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
41
|
Progressive striatonigral degeneration in a transgenic mouse model of multiple system atrophy: translational implications for interventional therapies. Acta Neuropathol Commun 2018; 6:2. [PMID: 29298733 PMCID: PMC5753576 DOI: 10.1186/s40478-017-0504-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/16/2017] [Indexed: 12/31/2022] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disorder characterized by widespread oligodendroglial cytoplasmic inclusions of filamentous α-synuclein, and neuronal loss in autonomic centres, basal ganglia and cerebellar circuits. It has been suggested that primary oligodendroglial α-synucleinopathy may represent a trigger in the pathogenesis of MSA, but the mechanisms underlying selective vulnerability and disease progression are unclear. The post-mortem analysis of MSA brains provides a static final picture of the disease neuropathology, but gives no clear indication on the sequence of pathogenic events in MSA. Therefore, alternative methods are needed to address these issues. We investigated selective vulnerability and disease progression in the transgenic PLP-α-syn mouse model of MSA characterized by targeted oligodendroglial α-synuclein overexpression aiming to provide a neuropathological correlate of motor deterioration. We show progressive motor deficits that emerge at 6 months of age and deteriorate up to 18 months of follow-up. The motor phenotype was associated with dopaminergic cell loss in the substantia nigra pars compacta at 6 months, followed by loss of striatal dopaminergic terminals and DARPP32-positive medium sized projection neurons at 12 months. Olivopontocerebellar motor loops remained spared in the PLP-α-syn model of MSA. These findings replicate progressive striatonigral degeneration underlying Parkinson-variant MSA. The initiation of the degenerative process was linked to an increase of soluble oligomeric α-synuclein species between 2 and 6 months. Early region-specific α-synuclein-associated activation profile of microglia was found in MSA substantia nigra. The role of abnormal neuroinflammatory signalling in disease progression was further supported by increased levels of CD68, CCL3, CCL5 and M-CSF with a peak in aged PLP-α-syn mice. In summary, transgenic PLP-α-syn mice show a distinctive oligodendroglial α-synucleinopathy that is associated with progressive striatonigral degeneration linked to abnormal neuroinflammatory response. The model provides a relevant tool for preclinical therapeutic target discovery for human Parkinson-variant MSA.
Collapse
|
42
|
Abstract
Fibromyalgia appears to present in subgroups with regard to biological pain induction, with primarily inflammatory, neuropathic/neurodegenerative, sympathetic, oxidative, nitrosative, or muscular factors and/or central sensitization. Recent research has also discussed glial activation or interrupted dopaminergic neurotransmission, as well as increased skin mast cells and mitochondrial dysfunction. Therapy is difficult, and the treatment options used so far mostly just have the potential to address only one of these aspects. As ambroxol addresses all of them in a single substance and furthermore also reduces visceral hypersensitivity, in fibromyalgia existing as irritable bowel syndrome or chronic bladder pain, it should be systematically investigated for this purpose. Encouraged by first clinical observations of two working groups using topical or oral ambroxol for fibromyalgia treatments, the present paper outlines the scientific argument for this approach by looking at each of the aforementioned aspects of this complex disease and summarizes putative modes of action of ambroxol. Nevertheless, at this point the evidence basis for ambroxol is not strong enough for clinical recommendation.
Collapse
Affiliation(s)
- Kai-Uwe Kern
- Institute of Pain Medicine/Pain Practice, Wiesbaden, Germany
| | | |
Collapse
|
43
|
Inhibition of the JAK/STAT Pathway Protects Against α-Synuclein-Induced Neuroinflammation and Dopaminergic Neurodegeneration. J Neurosci 2017; 36:5144-59. [PMID: 27147665 DOI: 10.1523/jneurosci.4658-15.2016] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/22/2016] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Parkinson's Disease (PD) is an age-related, chronic neurodegenerative disorder. At present, there are no disease-modifying therapies to prevent PD progression. Activated microglia and neuroinflammation are associated with the pathogenesis and progression of PD. Accumulation of α-synuclein (α-SYN) in the brain is a core feature of PD and leads to microglial activation, inflammatory cytokine/chemokine production, and ultimately to neurodegeneration. Given the importance of the JAK/STAT pathway in activating microglia and inducing cytokine/chemokine expression, we investigated the therapeutic potential of inhibiting the JAK/STAT pathway using the JAK1/2 inhibitor, AZD1480. In vitro, α-SYN exposure activated the JAK/STAT pathway in microglia and macrophages, and treatment with AZD1480 inhibited α-SYN-induced major histocompatibility complex Class II and inflammatory gene expression in microglia and macrophages by reducing STAT1 and STAT3 activation. For in vivo studies, we used a rat model of PD induced by viral overexpression of α-SYN. AZD1480 treatment inhibited α-SYN-induced neuroinflammation by suppressing microglial activation, macrophage and CD4(+) T-cell infiltration and production of proinflammatory cytokines/chemokines. Numerous genes involved in cell-cell signaling, nervous system development and function, inflammatory diseases/processes, and neurological diseases are enhanced in the substantia nigra of rats with α-SYN overexpression, and inhibited upon treatment with AZD1480. Importantly, inhibition of the JAK/STAT pathway prevented the degeneration of dopaminergic neurons in vivo These results indicate that inhibiting the JAK/STAT pathway can prevent neuroinflammation and neurodegeneration by suppressing activation of innate and adaptive immune responses to α-SYN. Furthermore, this suggests the feasibility of targeting the JAK/STAT pathway as a neuroprotective therapy for neurodegenerative diseases. SIGNIFICANCE STATEMENT α-SYN plays a central role in the pathophysiology of PD through initiation of neuroinflammatory responses. Using an α-SYN overexpression PD model, we demonstrate a beneficial therapeutic effect of AZD1480, a specific inhibitor of JAK1/2, in suppressing neuroinflammation and neurodegeneration. Our findings document that inhibition of the JAK/STAT pathway influences both innate and adaptive immune responses by suppressing α-SYN-induced microglia and macrophage activation and CD4(+) T-cell recruitment into the CNS, ultimately suppressing neurodegeneration. These findings are the first documentation that suppression of the JAK/STAT pathway disrupts the circuitry of neuroinflammation and neurodegeneration, thus attenuating PD pathogenesis. JAK inhibitors may be a viable therapeutic option for the treatment of PD patients.
Collapse
|
44
|
Joers V, Tansey MG, Mulas G, Carta AR. Microglial phenotypes in Parkinson's disease and animal models of the disease. Prog Neurobiol 2017; 155:57-75. [PMID: 27107797 PMCID: PMC5073045 DOI: 10.1016/j.pneurobio.2016.04.006] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/19/2022]
Abstract
Over the last decade the important concept has emerged that microglia, similar to other tissue macrophages, assume different phenotypes and serve several effector functions, generating the theory that activated microglia can be organized by their pro-inflammatory or anti-inflammatory and repairing functions. Importantly, microglia exist in a heterogenous population and their phenotypes are not permanently polarized into two categories; they exist along a continuum where they acquire different profiles based on their local environment. In Parkinson's disease (PD), neuroinflammation and microglia activation are considered neuropathological hallmarks, however their precise role in relation to disease progression is not clear, yet represent a critical challenge in the search of disease-modifying strategies. This review will critically address current knowledge on the activation states of microglia as well as microglial phenotypes found in PD and in animal models of PD, focusing on the expression of surface molecules as well as pro-inflammatory and anti-inflammatory cytokine production during the disease process. While human studies have reported an elevation of both pro- or anti-inflammatory markers in the serum and CSF of PD patients, animal models have provided insights on dynamic changes of microglia phenotypes in relation to disease progression especially prior to the development of motor deficits. We also review recent evidence of malfunction at multiple steps of NFκB signaling that may have a causal interrelationship with pathological microglia activation in animal models of PD. Finally, we discuss the immune-modifying strategies that have been explored regarding mechanisms of chronic microglial activation.
Collapse
Affiliation(s)
- Valerie Joers
- Department of Physiology, Emory University, Atlanta, GA, United States; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Malú G Tansey
- Department of Physiology, Emory University, Atlanta, GA, United States.
| | - Giovanna Mulas
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Italy.
| |
Collapse
|
45
|
Sanjari Moghaddam H, Zare-Shahabadi A, Rahmani F, Rezaei N. Neurotransmission systems in Parkinson’s disease. Rev Neurosci 2017; 28:509-536. [DOI: 10.1515/revneuro-2016-0068] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/10/2017] [Indexed: 12/17/2022]
Abstract
AbstractParkinson’s disease (PD) is histologically characterized by the accumulation of α-synuclein particles, known as Lewy bodies. The second most common neurodegenerative disorder, PD is widely known because of the typical motor manifestations of active tremor, rigidity, and postural instability, while several prodromal non-motor symptoms including REM sleep behavior disorders, depression, autonomic disturbances, and cognitive decline are being more extensively recognized. Motor symptoms most commonly arise from synucleinopathy of nigrostriatal pathway. Glutamatergic, γ-aminobutyric acid (GABA)ergic, cholinergic, serotoninergic, and endocannabinoid neurotransmission systems are not spared from the global cerebral neurodegenerative assault. Wide intrabasal and extrabasal of the basal ganglia provide enough justification to evaluate network circuits disturbance of these neurotransmission systems in PD. In this comprehensive review, English literature in PubMed, Science direct, EMBASE, and Web of Science databases were perused. Characteristics of dopaminergic and non-dopaminergic systems, disturbance of these neurotransmitter systems in the pathophysiology of PD, and their treatment applications are discussed.
Collapse
Affiliation(s)
- Hossein Sanjari Moghaddam
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Student Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Zare-Shahabadi
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Rahmani
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1419783151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Boston, MA, USA
| |
Collapse
|
46
|
Subramaniam SR, Federoff HJ. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson's Disease. Front Aging Neurosci 2017. [PMID: 28642697 PMCID: PMC5463358 DOI: 10.3389/fnagi.2017.00176] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Parkinson’s disease (PD) is a chronic and progressive disorder characterized neuropathologically by loss of dopamine neurons in the substantia nigra, intracellular proteinaceous inclusions, reduction of dopaminergic terminals in the striatum, and increased neuroinflammatory cells. The consequent reduction of dopamine in the basal ganglia results in the classical parkinsonian motor phenotype. A growing body of evidence suggest that neuroinflammation mediated by microglia, the resident macrophage-like immune cells in the brain, play a contributory role in PD pathogenesis. Microglia participate in both physiological and pathological conditions. In the former, microglia restore the integrity of the central nervous system and, in the latter, they promote disease progression. Microglia acquire different activation states to modulate these cellular functions. Upon activation to the M1 phenotype, microglia elaborate pro-inflammatory cytokines and neurotoxic molecules promoting inflammation and cytotoxic responses. In contrast, when adopting the M2 phenotype microglia secrete anti-inflammatory gene products and trophic factors that promote repair, regeneration, and restore homeostasis. Relatively little is known about the different microglial activation states in PD and a better understanding is essential for developing putative neuroprotective agents. Targeting microglial activation states by suppressing their deleterious pro-inflammatory neurotoxicity and/or simultaneously enhancing their beneficial anti-inflammatory protective functions appear as a valid therapeutic approach for PD treatment. In this review, we summarize microglial functions and, their dual neurotoxic and neuroprotective role in PD. We also review molecules that modulate microglial activation states as a therapeutic option for PD treatment.
Collapse
Affiliation(s)
| | - Howard J Federoff
- Department of Neurology, University of California, Irvine, Irvine, CAUnited States
| |
Collapse
|
47
|
Microglia in the primate macula: specializations in microglial distribution and morphology with retinal position and with aging. Brain Struct Funct 2017; 222:2759-2771. [PMID: 28213784 DOI: 10.1007/s00429-017-1370-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/16/2017] [Indexed: 01/08/2023]
Abstract
Microglia, the principal resident immune cell in the retina, play constitutive roles in immune surveillance and synapse maintenance, and are also associated with retinal disease, including those occurring in the macula. Perspectives on retinal microglia function have derived largely from rodent models and how these relate to the macula-bearing primate retina is unclear. In this study, we examined microglial distribution and cellular morphology in the adult rhesus macaque retina, and performed comparative characterizations in three retinal locations along the center-to-periphery axis (parafoveal, macular, and the peripheral retina). We found that microglia density peaked in the parafoveal retina and decreased in the peripheral retina. Individual microglial morphology reflected macular specialization, with macular microglia demonstrating the largest and most complex dendritic arbors relative to other retinal locations. Comparing retinal microglia between young and middle-aged animals, microglial density increased in the macular, but not in the peripheral retina with age, while microglial morphology across all locations remained relatively unchanged. Our findings indicate that microglial distribution and morphology demonstrate regional specialization in the retina, correlating with gradients of other retinal cell types. As microglia are innate immune cells implicated in age-related macular diseases, age-related microglial changes may be related to the increased vulnerability of the aged macula to immune-related neurodegeneration.
Collapse
|
48
|
Stayte S, Rentsch P, Tröscher AR, Bamberger M, Li KM, Vissel B. Activin A Inhibits MPTP and LPS-Induced Increases in Inflammatory Cell Populations and Loss of Dopamine Neurons in the Mouse Midbrain In Vivo. PLoS One 2017; 12:e0167211. [PMID: 28121982 PMCID: PMC5266209 DOI: 10.1371/journal.pone.0167211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023] Open
Abstract
Parkinson’s disease is a chronic neurodegenerative disease characterized by a significant loss of dopaminergic neurons within the substantia nigra pars compacta region and a subsequent loss of dopamine within the striatum. A promising avenue of research has been the administration of growth factors to promote the survival of remaining midbrain neurons, although the mechanism by which they provide neuroprotection is not understood. Activin A, a member of the transforming growth factor β superfamily, has been shown to be a potent anti-inflammatory following acute brain injury and has been demonstrated to play a role in the neuroprotection of midbrain neurons against MPP+-induced degeneration in vitro. We hypothesized that activin A may offer similar anti-inflammatory and neuroprotective effects in in vivo mouse models of Parkinson’s disease. We found that activin A significantly attenuated the inflammatory response induced by both MPTP and intranigral administration of lipopolysaccharide in C57BL/6 mice. We found that administration of activin A promoted survival of dopaminergic and total neuron populations in the pars compacta region both 8 days and 8 weeks after MPTP-induced degeneration. Surprisingly, no corresponding protection of striatal dopamine levels was found. Furthermore, activin A failed to protect against loss of striatal dopamine transporter expression in the striatum, suggesting the neuroprotective action of activin A may be localized to the substantia nigra. Together, these results provide the first evidence that activin A exerts potent neuroprotection and anti-inflammatory effects in the MPTP and lipopolysaccharide mouse models of Parkinson’s disease.
Collapse
Affiliation(s)
- Sandy Stayte
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Peggy Rentsch
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | - Kong M. Li
- Pharmacology Department, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Bryce Vissel
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
49
|
Fischer DL, Gombash SE, Kemp CJ, Manfredsson FP, Polinski NK, Duffy MF, Sortwell CE. Viral Vector-Based Modeling of Neurodegenerative Disorders: Parkinson's Disease. Methods Mol Biol 2016; 1382:367-82. [PMID: 26611600 DOI: 10.1007/978-1-4939-3271-9_26] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gene therapy methods are increasingly used to model Parkinson's disease (PD) in animals in an effort to test experimental therapeutics within a more relevant context to disease pathophysiology and neuropathology. We have detailed several criteria that are critical or advantageous to accurately modeling PD in a murine model or in a nonhuman primate. Using these criteria, we then evaluate approaches made to model PD using viral vectors to date, including both adeno-associated viruses and lentiviruses. Lastly, we comment on the consideration of aging as a critical factor for modeling PD.
Collapse
Affiliation(s)
- D Luke Fischer
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- MD/PhD Program, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Sara E Gombash
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Christopher J Kemp
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Fredric P Manfredsson
- Translational Science and Molecular Medicine, Michigan State University, College of Human Science, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Nicole K Polinski
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Caryl E Sortwell
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA.
| |
Collapse
|
50
|
Kong Y, Liang X, Liu L, Zhang D, Wan C, Gan Z, Yuan L. High Throughput Sequencing Identifies MicroRNAs Mediating α-Synuclein Toxicity by Targeting Neuroactive-Ligand Receptor Interaction Pathway in Early Stage of Drosophila Parkinson's Disease Model. PLoS One 2015; 10:e0137432. [PMID: 26361355 PMCID: PMC4567341 DOI: 10.1371/journal.pone.0137432] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 08/17/2015] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD) is a prevalent neurodegenerative disorder with pathological features including death of dopaminergic neurons in the substantia nigra and intraneuronal accumulations of Lewy bodies. As the main component of Lewy bodies, α-synuclein is implicated in PD pathogenesis by aggregation into insoluble filaments. However, the detailed mechanisms underlying α-synuclein induced neurotoxicity in PD are still elusive. MicroRNAs are ~20nt small RNA molecules that fine-tune gene expression at posttranscriptional level. A plethora of miRNAs have been found to be dysregulated in the brain and blood cells of PD patients. Nevertheless, the detailed mechanisms and their in vivo functions in PD still need further investigation. By using Drosophila PD model expressing α-synuclein A30P, we examined brain miRNA expression with high-throughput small RNA sequencing technology. We found that five miRNAs (dme-miR-133-3p, dme-miR-137-3p, dme-miR-13b-3p, dme-miR-932-5p, dme-miR-1008-5p) were upregulated in PD flies. Among them, miR-13b, miR-133, miR-137 are brain enriched and highly conserved from Drosophila to humans. KEGG pathway analysis using DIANA miR-Path demonstrated that neuroactive-ligand receptor interaction pathway was most likely affected by these miRNAs. Interestingly, miR-137 was predicted to regulate most of the identified targets in this pathway, including dopamine receptor (DopR, D2R), γ-aminobutyric acid (GABA) receptor (GABA-B-R1, GABA-B-R3) and N-methyl-D-aspartate (NMDA) receptor (Nmdar2). The validation experiments showed that the expression of miR-137 and its targets was negatively correlated in PD flies. Further experiments using luciferase reporter assay confirmed that miR-137 could act on specific sites in 3’ UTR region of D2R, Nmdar2 and GABA-B-R3, which downregulated significantly in PD flies. Collectively, our findings indicate that α-synuclein could induce the dysregulation of miRNAs, which target neuroactive ligand-receptor interaction pathway in vivo. We believe it will help us further understand the contribution of miRNAs to α-synuclein neurotoxicity and provide new insights into the pathogenesis driving PD.
Collapse
Affiliation(s)
- Yan Kong
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu Province, 210009, China
- * E-mail: (YK); (LY)
| | - Xijun Liang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Lin Liu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Dongdong Zhang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, 210009, China
| | - Chao Wan
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, 210009, China
| | - Zhenji Gan
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Liudi Yuan
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu Province, 210009, China
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, 210009, China
- * E-mail: (YK); (LY)
| |
Collapse
|