1
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [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: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
2
|
Shen T, Cui G, Chen H, Huang L, Song W, Zu J, Zhang W, Xu C, Dong L, Zhang Y. TREM-1 mediates interaction between substantia nigra microglia and peripheral neutrophils. Neural Regen Res 2024; 19:1375-1384. [PMID: 37905888 PMCID: PMC11467918 DOI: 10.4103/1673-5374.385843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/22/2023] [Accepted: 05/29/2023] [Indexed: 11/02/2023] Open
Abstract
Microglia-mediated neuroinflammation is considered a pathological feature of Parkinson’s disease. Triggering receptor expressed on myeloid cell-1 (TREM-1) can amplify the inherent immune response, and crucially, regulate inflammation. In this study, we found marked elevation of serum soluble TREM-1 in patients with Parkinson’s disease that positively correlated with Parkinson’s disease severity and dyskinesia. In a mouse model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson’s disease, we found that microglial TREM-1 expression also increased in the substantia nigra. Further, TREM-1 knockout alleviated dyskinesia in a mouse model of Parkinson’s disease and reduced dopaminergic neuronal injury. Meanwhile, TREM-1 knockout attenuated the neuroinflammatory response, dopaminergic neuronal injury, and neutrophil migration. Next, we established an in vitro 1-methyl-4-phenyl-pyridine-induced BV2 microglia model of Parkinson’s disease and treated the cells with the TREM-1 inhibitory peptide LP17. We found that LP17 treatment reduced apoptosis of dopaminergic neurons and neutrophil migration. Moreover, inhibition of neutrophil TREM-1 activation diminished dopaminergic neuronal apoptosis induced by lipopolysaccharide. TREM-1 can activate the downstream CARD9/NF-κB proinflammatory pathway via interaction with SYK. These findings suggest that TREM-1 may play a key role in mediating the damage to dopaminergic neurons in Parkinson’s disease by regulating the interaction between microglia and peripheral neutrophils.
Collapse
Affiliation(s)
- Tong Shen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hao Chen
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Long Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| | - Wei Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wei Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chuanying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Liguo Dong
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yongmei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| |
Collapse
|
3
|
Dzamko N. Cytokine activity in Parkinson's disease. Neuronal Signal 2023; 7:NS20220063. [PMID: 38059210 PMCID: PMC10695743 DOI: 10.1042/ns20220063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023] Open
Abstract
The contribution of the immune system to the pathophysiology of neurodegenerative Parkinson's disease (PD) is increasingly being recognised, with alterations in the innate and adaptive arms of the immune system underlying central and peripheral inflammation in PD. As chief modulators of the immune response, cytokines have been intensely studied in the field of PD both in terms of trying to understand their contribution to disease pathogenesis, and if they may comprise much needed therapeutic targets for a disease with no current modifying therapy. This review summarises current knowledge on key cytokines implicated in PD (TNFα, IL-6, IL-1β, IL-10, IL-4 and IL-1RA) that can modulate both pro-inflammatory and anti-inflammatory effects. Cytokine activity in PD is clearly a complicated process mediated by substantial cross-talk of signalling pathways and the need to balance pro- and anti-inflammatory effects. However, understanding cytokine activity may hold promise for unlocking new insight into PD and how it may be halted.
Collapse
Affiliation(s)
- Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine and Health and the Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| |
Collapse
|
4
|
Contaldi E, Magistrelli L, Comi C. Disease mechanisms as subtypes: Immune dysfunction in Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:67-93. [PMID: 36803824 DOI: 10.1016/b978-0-323-85555-6.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In recent years, the contraposition between inflammatory and neurodegenerative processes has been increasingly challenged. Inflammation has been emphasized as a key player in the onset and progression of Parkinson disease (PD) and other neurodegenerative disorders. The strongest indicators of the involvement of the immune system derived from evidence of microglial activation, profound imbalance in phenotype and composition of peripheral immune cells, and impaired humoral immune responses. Moreover, peripheral inflammatory mechanisms (e.g., involving the gut-brain axis) and immunogenetic factors are likely to be implicated. Even though several lines of preclinical and clinical studies are supporting and defining the complex relationship between the immune system and PD, the exact mechanisms are currently unknown. Similarly, the temporal and causal connections between innate and adaptive immune responses and neurodegeneration are unsettled, challenging our ambition to define an integrated and holistic model of the disease. Despite these difficulties, current evidence is providing the unique opportunity to develop immune-targeted approaches for PD, thus enriching our therapeutic armamentarium. This chapter aims to provide an extensive overview of past and present studies that explored the implication of the immune system in neurodegeneration, thus paving the road for the concept of disease modification in PD.
Collapse
Affiliation(s)
- Elena Contaldi
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Luca Magistrelli
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Cristoforo Comi
- Neurology Unit, S.Andrea Hospital, Department of Translational Medicine, University of Piemonte Orientale, Vercelli, Italy.
| |
Collapse
|
5
|
Wu Q, Wang M, Chen W, Wang K, Wang Y. Assessing neuroprotective efficacy of phytochemical saponin ruscogenin in both in vitro and in vivo model. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
|
6
|
Li S, Bi G, Han S, Huang R. MicroRNAs Play a Role in Parkinson’s Disease by Regulating Microglia Function: From Pathogenetic Involvement to Therapeutic Potential. Front Mol Neurosci 2022; 14:744942. [PMID: 35126050 PMCID: PMC8814625 DOI: 10.3389/fnmol.2021.744942] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) is a clinically common neurodegenerative disease of the central nervous system (CNS) characterized by loss of dopamine neurons in the substantia nigra. Microglia (MG), as an innate immune cell in the CNS, are involved in a variety of immunity and inflammatory responses in the CNS. A number of studies have shown that the overactivation of MG is one of the critical pathophysiological mechanisms underlying PD. MicroRNAs (miRNAs) are considered to be an important class of gene expression regulators and are involved in a variety of physiological and pathological mechanisms, including immunity and inflammation. In addition, miRNAs can affect the progress of PD by regulating the expression of various MG genes and the polarization state of the MG. Here, we summarize recent articles and describe the important role of MG pathological polarization in the progression of PD, the diverse mechanisms responsible for how miRNAs regulate MG, and the potential therapeutic prospects of miRNAs for PD. We also propose that the regulation of miRNAs may be a novel protective approach against the pathogenesis of PD.
Collapse
Affiliation(s)
- Silu Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guorong Bi
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shunchang Han
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Rui Huang,
| |
Collapse
|
7
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
8
|
Bagheri-Mohammadi S. Stem cell-based therapy as a promising approach in Alzheimer's disease: current perspectives on novel treatment. Cell Tissue Bank 2021; 22:339-353. [PMID: 33398492 DOI: 10.1007/s10561-020-09896-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neuronal disorder with insidious onset and slow progression, leading to growing global concern with huge implications for individuals and society. The occurrence of AD has been increased and has become an important health issue throughout the world. In recent years, the care of more than 35 million patients with AD costs over $ 600 billion per year, it is approximately 1 percent of the global Gross Domestic Product. Currently, the therapeutic approach is not effective for neurological deficits especially after the development of these major neurological disorders. The discovery of the technique called cell-based therapy has shown promising results and made important conclusions beyond AD using the stem cells approach. Here we review recent progress on stem cell-based therapy in the context of AD.
Collapse
Affiliation(s)
- Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
9
|
Jones M, Nankervis B, Roballo KS, Pham H, Bushman J, Coeshott C. A Comparison of Automated Perfusion- and Manual Diffusion-Based Human Regulatory T Cell Expansion and Functionality Using a Soluble Activator Complex. Cell Transplant 2021; 29:963689720923578. [PMID: 32662685 PMCID: PMC7586259 DOI: 10.1177/0963689720923578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Absence or reduced frequency of human regulatory T cells (Tregs) can limit the control of inflammatory responses, autoimmunity, and the success of transplant engraftment. Clinical studies indicate that use of Tregs as immunotherapeutics would require billions of cells per dose. The Quantum® Cell Expansion System (Quantum system) is a hollow-fiber bioreactor that has previously been used to grow billions of functional T cells in a short timeframe, 8–9 d. Here we evaluated expansion of selected Tregs in the Quantum system using a soluble activator to compare the effects of automated perfusion with manual diffusion-based culture in flasks. Treg CD4+CD25+ cells from three healthy donors, isolated via column-free immunomagnetic negative/positive selection, were grown under static conditions and subsequently seeded into Quantum system bioreactors and into T225 control flasks in an identical culture volume of PRIME-XV XSFM medium with interleukin-2, for a 9-d expansion using a soluble anti-CD3/CD28/CD2 monoclonal antibody activator complex. Treg harvests from three parallel expansions produced a mean of 3.95 × 108 (range 1.92 × 108 to 5.58 × 108) Tregs in flasks (mean viability 71.3%) versus 7.00 × 109 (range 3.57 × 109 to 13.00 × 109) Tregs in the Quantum system (mean viability 91.8%), demonstrating a mean 17.7-fold increase in Treg yield for the Quantum system over that obtained in flasks. The two culture processes gave rise to cells with a memory Treg CD4+CD25+FoxP3+CD45RO+ phenotype of 93.7% for flasks versus 97.7% for the Quantum system. Tregs from the Quantum system demonstrated an 8-fold greater interleukin-10 stimulation index than cells from flask culture following restimulation. Quantum system–expanded Tregs proliferated, maintained their antigenic phenotype, and suppressed effector immune cells after cryopreservation. We conclude that an automated perfusion bioreactor can support the scale-up expansion of functional Tregs more efficiently than diffusion-based flask culture.
Collapse
Affiliation(s)
| | | | | | - Huong Pham
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Jared Bushman
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | | |
Collapse
|
10
|
Burtscher J, Millet GP, Place N, Kayser B, Zanou N. The Muscle-Brain Axis and Neurodegenerative Diseases: The Key Role of Mitochondria in Exercise-Induced Neuroprotection. Int J Mol Sci 2021; 22:6479. [PMID: 34204228 PMCID: PMC8235687 DOI: 10.3390/ijms22126479] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Regular exercise is associated with pronounced health benefits. The molecular processes involved in physiological adaptations to exercise are best understood in skeletal muscle. Enhanced mitochondrial functions in muscle are central to exercise-induced adaptations. However, regular exercise also benefits the brain and is a major protective factor against neurodegenerative diseases, such as the most common age-related form of dementia, Alzheimer's disease, or the most common neurodegenerative motor disorder, Parkinson's disease. While there is evidence that exercise induces signalling from skeletal muscle to the brain, the mechanistic understanding of the crosstalk along the muscle-brain axis is incompletely understood. Mitochondria in both organs, however, seem to be central players. Here, we provide an overview on the central role of mitochondria in exercise-induced communication routes from muscle to the brain. These routes include circulating factors, such as myokines, the release of which often depends on mitochondria, and possibly direct mitochondrial transfer. On this basis, we examine the reported effects of different modes of exercise on mitochondrial features and highlight their expected benefits with regard to neurodegeneration prevention or mitigation. In addition, knowledge gaps in our current understanding related to the muscle-brain axis in neurodegenerative diseases are outlined.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Grégoire P. Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Bengt Kayser
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| |
Collapse
|
11
|
Adeno-Associated Viral Vectors as Versatile Tools for Parkinson's Research, Both for Disease Modeling Purposes and for Therapeutic Uses. Int J Mol Sci 2021; 22:ijms22126389. [PMID: 34203739 PMCID: PMC8232322 DOI: 10.3390/ijms22126389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022] Open
Abstract
It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson’s disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson’s disease and related synucleinopathies.
Collapse
|
12
|
Bonte MA, El Idrissi F, Gressier B, Devos D, Belarbi K. Protein network exploration prioritizes targets for modulating neuroinflammation in Parkinson's disease. Int Immunopharmacol 2021; 95:107526. [PMID: 33756233 DOI: 10.1016/j.intimp.2021.107526] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/20/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disease associated with a loss of dopaminergic neurons in the substantia nigra of the brain. Neuroinflammation, another hallmark of the disease, is thought to play an important role in the neurodegenerative process. While mitigating neuroinflammation could prove beneficial for Parkinson's disease, identifying the most relevant biological processes and pharmacological targets as well as drugs to modulate them remains highly challenging. The present study aimed to better understand the protein network behind neuroinflammation in Parkinson's disease and to prioritize possible targets for its pharmacological modulation. We first used text-mining to systematically collect the proteins significantly associated to Parkinson's disease neuroinflammation over the scientific literature. The functional interaction network formed by these proteins was then analyzed by integrating functional enrichment, network topology analysis and drug-protein interaction analysis. We identified 57 proteins significantly associated to neuroinflammation in Parkinson's disease. Toll-like Receptor Cascades as well as Interleukin 4, Interleukin 10 and Interleukin 13 signaling appeared as the most significantly enriched biological processes. Protein network analysis using STRING and CentiScaPe identified 8 proteins with the highest ability to control these biological processes underlying neuroinflammation, namely caspase 1, heme oxygenase 1, interleukin 1beta, interleukin 4, interleukin 6, interleukin 10, tumor necrosis factor alpha and toll-like receptor 4. These key proteins were indexed to be targetable by a total of 38 drugs including 27 small compounds 11 protein-based therapies. In conclusion, our study highlights key proteins in Parkinson's disease neuroinflammation as well as pharmacological compounds acting on them. As such, it may facilitate the prioritization of biomarkers for the development of diagnostic, target-engagement assessment and therapeutic tools against Parkinson's disease.
Collapse
Affiliation(s)
- Marie-Amandine Bonte
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France.
| | - Fatima El Idrissi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| | - Bernard Gressier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie Médicale, I-SITE ULNE, LiCEND, Lille, France.
| | - Karim Belarbi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, F-59000 Lille, France; Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France.
| |
Collapse
|
13
|
Jiao L, Du X, Jia F, Li Y, Zhu D, Tang T, Jiao Q, Jiang H. Early low-dose ghrelin intervention via miniosmotic pumps could protect against the progressive dopaminergic neuron loss in Parkinson's disease mice. Neurobiol Aging 2021; 101:70-78. [PMID: 33582568 DOI: 10.1016/j.neurobiolaging.2021.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 01/08/2023]
Abstract
Ghrelin has been identified as a multifunctional peptide that has a potential application for treating Parkinson's disease (PD). The objective of this study was to assess the effects of subcutaneous administration of low-dose ghrelin via miniosmotic pumps on PD progression. The decreased levels of total and active ghrelin in plasma were rescued by ghrelin administration in PD mice. Interestingly, ghrelin did not affect weight gain in wild-type mice but improved weight loss in PD mice. We observed the attenuation of dopaminergic neuron loss in substantia nigra and a low level of dopamine content in the striatum in PD mice with ghrelin treatment. Ghrelin administration could improve the microenvironment of dopaminergic neurons by inhibiting microglial proliferation and proinflammatory cytokine expression and could enhance cell survival by upregulating Bcl-2/Bax ratio and superoxide dismutase1 protein level in the substantia nigra of PD mice. Subcutaneous administration of low-dose ghrelin could prevent the onset of the progression of PD and also provide a possible method for ghrelin application to cure PD.
Collapse
Affiliation(s)
- Lingling Jiao
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Fengju Jia
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Yong Li
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Dexiao Zhu
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Tinging Tang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China.
| |
Collapse
|
14
|
Chen Y, Zhu G, Liu D, Zhang X, Liu Y, Yuan T, Du T, Zhang J. Subthalamic nucleus deep brain stimulation suppresses neuroinflammation by Fractalkine pathway in Parkinson's disease rat model. Brain Behav Immun 2020; 90:16-25. [PMID: 32726685 DOI: 10.1016/j.bbi.2020.07.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 11/24/2022] Open
Abstract
Subthalamic nucleus deep brain stimulation (STN-DBS) is widely used to treat patients with Parkinson's disease (PD), and recent studies have shown that it is more beneficial for early stages, suggesting a potential neuroprotective effect. And the neuroinflammation plays an indispensable role in progress of PD. However, the underlying mechanisms are not well understood. The aim of this study was to investigate the effect of STN-DBS on neuroinflammation and the potential pathway. To address this question, we established a rat PD model by unilateral 6-hydroxydopamine injection into the left striatum and implanted stimulation leads into the ipsilateral STN to deliver electrical stimulation for a week. The neuroprotective effects of STN-DBS were examined by molecular biology techniques, including western blotting, immunohistochemistry and so on. We found that motor deficits were alleviated by STN-DBS, with increased survival of dopaminergic neurons in the substantia nigra (SN). Furthermore, STN-DBS decreased Fractalkine (CX3CL1) and its receptor (CX3CR1) expression. Meanwhile, the suppressed microglia activation and nuclear factor-κB expression, decrease in the levels of pro-inflammatory cytokine interleukin (IL)-1β and IL-6 and increase in anti-inflammatory cytokine IL-4, downregulated IL-1 receptor, extracellular signal-regulated kinase (ERK) and cleaved-caspase3 were also observed in SN of PD models received STN-DBS. In conclusion, we observed a significant association between the suppressed neuroinflammation and STN-DBS, which may be attributed to CX3CL1/CX3CR1 signaling. These results provide novel insight into the mechanistic basis of STN-DBS therapy for PD.
Collapse
Affiliation(s)
- Yingchuan Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Guanyu Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Defeng Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xin Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Yuye Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Tianshuo Yuan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Tingting Du
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China; Beijing Key Laboratory of Neurostimulation, Beijing 100070, China.
| |
Collapse
|
15
|
Low frequency electroacupuncture alleviates neuropathic pain by activation of spinal microglial IL-10/β-endorphin pathway. Biomed Pharmacother 2020; 125:109898. [DOI: 10.1016/j.biopha.2020.109898] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/01/2020] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
|
16
|
Dwyer Z, Rudyk C, Situt D, Beauchamp S, Abdali J, Dinesh A, Legancher N, Sun H, Schlossmacher M, Hayley S. Microglia depletion prior to lipopolysaccharide and paraquat treatment differentially modulates behavioral and neuronal outcomes in wild type and G2019S LRRK2 knock-in mice. Brain Behav Immun Health 2020; 5:100079. [PMID: 34589856 PMCID: PMC8474533 DOI: 10.1016/j.bbih.2020.100079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Substantial data have implicated microglial-driven neuroinflammation in Parkinson's disease (PD) and environmental toxicants have been long expected as triggers of such inflammatory processes. Of course, these environmental insults act in the context of genetic vulnerability factors and in this regard, leucine rich repeat kinase 2 (LRRK2), may play a prominent role. METHODS We used a double hit, lipopolysaccharide (LPS; endotoxin) followed by paraquat (pesticide toxicant) model of PD in mice with the most common LRRK2 mutation G2019S, knockin mice and wild type littermates. In order to assess the contribution of microglia, we depleted these cells (through 14 days of the CSF-1 antagonist, PLX-3397) prior to LPS and paraquat exposure. RESULTS We found that the G2019S mice displayed the greatest signs of behavioral pathology, but that the PLX-3397 induced microglial depletion at the time of LPS exposure diminished toxicity and weight loss and blunted the reduction in home-cage activity with subsequent paraquat exposure. However, neither the PLX-3397 pre-treatment nor the G2019S mutation affected the LPS + paraquat induced loss of substantia nigra pars compacta (SNc) dopamine neurons or elevation of circulating immune (IL-6) or stress (corticosterone) factors. Intriguingly, microglial morphological ratings were basally enhanced in G2019S mice and the PLX-3397 pre-treatment reversed this effect. Moreover, PLX-3397 pre-treatment selectively elevated soluble a-synuclein and SIRT3 levels, while reducing SNc caspase-1 and 3, along with CX3CR1. Hence, the re-populated "new" microglia following cessation of PLX-3397 clearly had an altered phenotype or were immature at the time of sacrifice (i.e. after 11 days). CONCLUSIONS Collectively, these findings suggest that G2019S knock-in and PLX-3397 microglial depletion at the time of LPS exposure affects behavioral, but not neurodegenerative responses to subsequent environmental toxin exposure.
Collapse
Affiliation(s)
- Zach Dwyer
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Chris Rudyk
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Divya Situt
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Sheryl Beauchamp
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Jawaria Abdali
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Anu Dinesh
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | | | - Hongyu Sun
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | | | - Shawn Hayley
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - CLINT (Canadian LRRK2 in inflammation team)
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada
- University of Ottawa, Canada
- Ottawa Hospital Research Institute, Canada
| |
Collapse
|
17
|
Ulhaq ZS, Garcia CP. Inflammation-related gene polymorphisms associated with Parkinson’s disease: an updated meta-analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2020. [DOI: 10.1186/s43042-020-00056-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Background
Strong evidence supports the involvement of inflammation processes in the development and progression of Parkinson’s disease (PD), where increasingly correlations have been identified between genetic variations in inflammation-related genes and PD. However, data varies between studies. Therefore, we conducted a meta-analysis to clarify associations between inflammation-related gene polymorphisms and PD risk.
Methods
All studies were identified through online databases. Pooled and stratified groups based on racial descent were assembled to evaluate associations between polymorphisms and PD.
Results
The pooled results showed that protective effects for PD were observed for (1) IL-1α -889 C/T in Asian populations (T vs. C, OR = 0.831, P = 0.031; TT + CT vs. CC, OR = 0.827, P = 0.049); (2) IL-6 -176 G/C in Caucasian populations (CC + GC vs. GG, OR = 0.656, P = 0.000; GC vs. GG, OR = 0.673, P = 0.000); (3) IL-8 -251 A/T (T vs. A, OR = 0.812, P = 0.041; TT vs. AT + AA, OR = 0.663, P = 0.012), particularly in Caucasian populations (TT vs. AT + AA, OR = 0.639, P = 0.010); (4) IL-10 -819 T/C (C vs. T, OR = 0.742, P = 0.034); (5) IL-18 -607 C/A (AA + CA vs. CC, OR = 0.597, P = 0.015; CA vs. CC, OR = 0.534, P = 0.005), and (6) CCR2 +190 G/A (AA vs. GA + GG, OR = 0.552, P = 0.018; AA vs. GG; OR = 0.554; 95% CI 0.336–0.914, P = 0.005). An increased risk of PD was associated with IL-10 -1082 G/A in Asian populations (A vs. G, OR = 1.731, P = 0.000; AA + GA vs. GG, OR = 1.910, P = 0.000). No significant associations with PD were observed for polymorphisms in IL-1β -511 C/T, IL-10 -592 C/A, IL-18 -137 G/C, TNFα -863 C/A, TNFα -857 C/T, TNFα -308 G/A, IFNΥ +874 T/A, and MCP1/CCL2 +2518 A/G.
Conclusions
We suggest that IL-1α -889, IL-6 -176, IL-8 -251, IL-10 -1082, IL-10 -819, IL-18 -607, and CCR2 +190 polymorphisms may be associated with PD risk; however, further studies must verify these conclusions.
Collapse
|
18
|
Gastrointestinal Dysfunctions Are Associated with IL-10 Variants in Parkinson's Disease. PARKINSONS DISEASE 2019; 2018:5908359. [PMID: 30631418 PMCID: PMC6304865 DOI: 10.1155/2018/5908359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 11/17/2022]
Abstract
Inflammation has been demonstrated to be involved in Parkinson's disease (PD) pathogenesis. There were evidences that the disturbance of the protective function of IL-10 gene contributed to PD. In our study, haplotype analyses were conducted of IL-10 rs1800871 and rs1800872 on 371 PD patients. Because the two SNPs exposed significant linkage disequilibrium demonstrated by Haploview software, we included 177 carriers of both rs1800871 and rs1800872 and 190 noncarriers in clinical phenotype analyses. As to nonmotor symptoms, the score of the gastrointestinal dysfunction domain in Nonmotor Symptom Scale (NMSS) was lower in the carrier group of both SNPs than in the noncarrier group in PD patients (SC: -0.198, p : 023). Other nonmotor symptoms reflected by relevant rating scales showed negative results. As to comorbidity, no significant statistical significance was observed between the two SNPs and Charlson Comorbidity Index (CCI). In conclusion, we found less severe gastrointestinal dysfunctions of both IL-10 rs1800871 and rs1800872 carriers than noncarriers in PD.
Collapse
|
19
|
Arce-Sillas A, Sevilla-Reyes E, Álvarez-Luquín DD, Guevara-Salinas A, Boll MC, Pérez-Correa CA, Vivas-Almazan AV, Rodríguez-Ortiz U, Castellanos Barba C, Hernandez M, Fragoso G, Sciutto E, Cárdenas G, Adalid-Peralta LV. Expression of Dopamine Receptors in Immune Regulatory Cells. Neuroimmunomodulation 2019; 26:159-166. [PMID: 31311029 DOI: 10.1159/000501187] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/28/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Parkinson's disease (PD) patients are usually treated with L-dopa and/or dopaminergic agonists, which act by binding five types of dopaminergic receptors (DRD1-DRD5). Peripheral immune cells are known to express dopamine receptors on their membrane surface, and therefore they could be directly affected by the treatment. Regulatory cells are the main modulators of inflammation, but it is not clear whether dopaminergic treatment could affect their functions. While only regulatory T cells (Tregs) have been proved to express dopamine receptors, it is not known whether other regulatory cells such as CD8regs, regulatory B cells (Bregs), tolerogenic dendritic cells, and intermediate monocytes also express them. METHODS The expression of dopamine receptors in Tregs, CD8regs, Bregs, tolerogenic dendritic cells, and intermediate monocytes was herein evaluated. cDNA from 11 PD patients and 9 control subjects was obtained and analyzed. RESULTS All regulatory cell populations expressed the genes coding for dopamine receptors, and this expression was further corroborated by flow cytometry. These findings may allow us to propose regulatory populations as possible targets for PD treatment. CONCLUSIONS This study opens new paths to deepen our understanding on the effect of PD treatment on the cells of the regulatory immune response.
Collapse
Affiliation(s)
- Asiel Arce-Sillas
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Edgar Sevilla-Reyes
- Clinica de investigación en enfermedades infecciosas (CIENI), Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Diana Denisse Álvarez-Luquín
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Adrian Guevara-Salinas
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | | | - Citzielli Aseret Pérez-Correa
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Alma Viridiana Vivas-Almazan
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | | | - Carlos Castellanos Barba
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marisela Hernandez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edda Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Graciela Cárdenas
- Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Laura Virginia Adalid-Peralta
- Unidad Periférica para el Estudio de la Neuroinflamación en Patologías Neurológicas, Instituto de Investigaciones Biomédicas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico,
- Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico,
| |
Collapse
|
20
|
Baicalin Exerts Anti-Airway Inflammation and Anti-Remodelling Effects in Severe Stage Rat Model of Chronic Obstructive Pulmonary Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:7591348. [PMID: 30402133 PMCID: PMC6196890 DOI: 10.1155/2018/7591348] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/06/2018] [Indexed: 12/16/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a worldwide epidemic. Current approaches are disappointing due to limited improvement of the disease development. The present study established 36-week side stream cigarette smoke induced rat model of COPD with advanced stage feature and evaluted the effects of baicalin on the model. Fifty-four Sprague–Dawley rats were randomly divided into six groups including room air control, cigarette smoke exposure, baicalin (40 mg/kg, 80 mg/kg, and 160 mg/kg), and budesonide used as a positive control. Rats were exposed to cigarette smoke from 3R4F research cigarettes. Pulmonary function was evaluated and pathological changes were also observed. Cytokine level related to airway inflammation and remodelling in blood serum, bronchoalveolar lavage fluid, and lung tissue was determined. Blood gases and HPA axis function were also examined, and antioxidant levels were quantified. Results showed that, after treatment with baicalin, lung function was improved and histopathological changes were ameliorated. Baicalin also regulated proinflammatory and anti-inflammatory balance and also airway remodelling and anti-airway remodelling factors in blood serum, bronchoalveolar lavage fluid, and lung tissue. Antioxidant capacity was also increased after treatment with baicalin in COPD rat model. HPA axis function was improved in baicalin treated groups as compared to model group. Therefore, baicalin exerts lung function protection, proinflammatory and anti-inflammatory cytokine regulation, anti-airway remodelling, and antioxidant role in long term CS induced COPD model.
Collapse
|
21
|
Sharma M, Flood PM. β-arrestin2 regulates the anti-inflammatory effects of Salmeterol in lipopolysaccharide-stimulated BV2 cells. J Neuroimmunol 2018; 325:10-19. [PMID: 30352316 DOI: 10.1016/j.jneuroim.2018.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Microglial activation contributes to chronic inflammation and neuronal loss in progressive neurodegenerative disorders such as Parkinson's disease (PD). Thus, treatments suppressing microglial activation may have therapeutic benefits to prevent neuronal loss in neurodegenerative diseases. Our previous findings show that Salmeterol, a long-acting β2-adrenergic receptor (β2-AR) agonist, is neuroprotective in two distinct animal models of PD, including where lipopolysaccharide (LPS) from E. coli was used to initiate chronic neurodegeneration. Salmeterol was found to be a potent inhibitor of dopaminergic neurodegeneration by regulating the production of pro-inflammatory mediators from activated microglial cells. In the present study, we investigated the molecular basis of the anti-inflammatory effects of Salmeterol on LPS-activated murine microglial BV2 cells. BV2 cells were pretreated with Salmeterol and followed by stimulation with LPS. Salmeterol inhibited LPS-induced release of the pro-inflammatory mediators such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and nitric oxide from BV2 cells. Additionally, Salmeterol suppressed nuclear translocation of nuclear factor kappa-B (NF-κB) p65 by inhibiting the IκB-α degradation and TAK1 (transforming growth factor-beta-activated kinase1) phosphorylation. We have also found that Salmeterol increases the expression of β-arrestin2 and enhances the interaction between β-arrestin2 and TAB1 (TAK1-binding protein), reduced TAK1/TAB1 mediated activation of NFκB and expression of pro-inflammatory genes. Furthermore, silencing of β-arrestin2 abrogates the anti-inflammatory effects of Salmeterol in LPS-stimulated BV2 cells. Our findings suggest that the anti-inflammatory properties of Salmeterol is β-arrestin2 dependent and also offers novel therapeutics targeting inflammatory pathways to prevent microglial cell activation and neuronal loss in neuroinflammatory diseases like PD.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Patrick M Flood
- Departments of Dentistry and Medical Microbiology and Immunology, and Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
22
|
Wu HY, Mao XF, Tang XQ, Ali U, Apryani E, Liu H, Li XY, Wang YX. Spinal interleukin-10 produces antinociception in neuropathy through microglial β-endorphin expression, separated from antineuroinflammation. Brain Behav Immun 2018; 73:504-519. [PMID: 29928964 DOI: 10.1016/j.bbi.2018.06.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin 10 (IL-10) is antinociceptive in various animal models of pain without induction of tolerance, and its mechanism of action was generally believed to be mediated by inhibition of neuroinflammation. Here we reported that intrathecal IL-10 injection dose dependently attenuated mechanical allodynia and thermal hyperalgesiain male and female neuropathic rats, with ED50 values of 40.8 ng and 24 ng, and Emax values of 61.5% MPE and 100% MPE in male rats. Treatment with IL-10 specifically increased expression of the β-endorphin (but not prodynorphin) gene and protein in primary cultures of spinal microglia but not in astrocytes or neurons. Intrathecal injection of IL-10 stimulated β-endorphin expression from microglia but not neurons or astrocytes in both contralateral and ipsilateral spinal cords of neuropathic rats. However, intrathecal injection of the β-endorphin neutralizing antibody, opioid receptor antagonist naloxone, or μ-opioid receptor antagonist CTAP completely blocked spinal IL-10-induced mechanical antiallodynia, while the microglial inhibitor minocycline and specific microglia depletor reversed spinal IL-10-induced β-endorphin overexpression and mechanical antiallodynia. IL-10 treatment increased spinal microglial STAT3 phosphorylation, and the STAT3 inhibitor NSC74859 completely reversed IL-10-increased spinal expression of β-endorphin and neuroinflammatory cytokines and mechanical antiallodynia. Silence of the Bcl3 and Socs3 genes nearly fully reversed IL-10-induced suppression of neuroinflammatory cytokines (but not expression of β-endorphin), although it had no effect on mechanical allodynia. In contrast, disruption of the POMC gene completely blocked IL-10-stimulated β-endorphin expression and mechanical antiallodynia, but had no effect on IL-10 inhibited expression of neuroinflammatory cytokines. Thus this study revealed that IL-10 produced antinociception through spinal microglial β-endorphin expression, but not inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Hai-Yun Wu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xiao-Fang Mao
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xue-Qi Tang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Usman Ali
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Evhy Apryani
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Hao Liu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xin-Yan Li
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China.
| |
Collapse
|
23
|
Saghazadeh A, Ferrari CC, Rezaei N. Deciphering variability in the role of interleukin-1β in Parkinson's disease. Rev Neurosci 2018; 27:635-50. [PMID: 27166719 DOI: 10.1515/revneuro-2015-0059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Although the role of inflammation in neurodegeneration has been well acknowledged, less is known on the issue of each cytokine in specific neurodegenerative diseases. In this review, we will present evidence elucidating that interleukin-1β (IL-1β) has a multi-faceted character in pathogenesis of Parkinson's disease, which is a progressive neurodegenerative disorder. Increased levels of IL-1β were found in PD patients. Besides, PD symptoms were observed in IL-1β wild-type, but not deficient, animals. These lines of evidence suggest that IL-1β may contribute to the initiation or progression of PD. On the other hand, some studies reported decreased levels of IL-1β in PD patients. Also, genetic studies provided evidence suggesting that IL-1β may protect individuals against PD. Presumably, the broad range of IL-1β role is due to its interaction with both upstream and downstream mediators. Differences in IL-1β levels could be because of glia population (i.e. microglia and astrocytes), mitogen-activated protein kinase and nuclear factor κ light-chain-enhancer of activated B cells signaling pathways, and several mediators (including cyclooxygenase, neurotrophic factors, reactive oxygen species, caspases, heme oxygenase-1, and matrix metalloproteinases). Although far from practice at this point, unraveling theoretical therapeutic targets based on the up-down IL-1β neuroweb could facilitate the development of strategies that are likely to be used for pharmaceutical designs of anti-neurodegenerative drugs of the future.
Collapse
|
24
|
Zhou X, Spittau B. Lipopolysaccharide-Induced Microglia Activation Promotes the Survival of Midbrain Dopaminergic Neurons In Vitro. Neurotox Res 2017; 33:856-867. [DOI: 10.1007/s12640-017-9842-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
|
25
|
Rocha Sobrinho HMD, Silva DJD, Gomides LF, Dorta ML, Oliveira MAPD, Ribeiro-Dias F. TLR4 and TLR2 activation is differentially associated with age during Parkinson’s disease. Immunol Invest 2017; 47:71-88. [DOI: 10.1080/08820139.2017.1379024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Delson José da Silva
- Hospital das Clínicas, Universidade Federal de Goiás and Instituto Integrado de Neurociências, Goiânia, Goiás, Brazil
| | - Larissa Fonseca Gomides
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Miriam Leandro Dorta
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | | | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| |
Collapse
|
26
|
Let's make microglia great again in neurodegenerative disorders. J Neural Transm (Vienna) 2017; 125:751-770. [PMID: 29027011 DOI: 10.1007/s00702-017-1792-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
All of the common neurodegenerative disorders-Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prion diseases-are characterized by accumulation of misfolded proteins that trigger activation of microglia; brain-resident mononuclear phagocytes. This chronic form of neuroinflammation is earmarked by increased release of myriad cytokines and chemokines in patient brains and biofluids. Microglial phagocytosis is compromised early in the disease process, obfuscating clearance of abnormal proteins. This review identifies immune pathologies shared by the major neurodegenerative disorders. The overarching concept is that aberrant innate immune pathways can be targeted for return to homeostasis in hopes of coaxing microglia into clearing neurotoxic misfolded proteins.
Collapse
|
27
|
Zhu Y, Liu Z, Peng YP, Qiu YH. Interleukin-10 inhibits neuroinflammation-mediated apoptosis of ventral mesencephalic neurons via JAK-STAT3 pathway. Int Immunopharmacol 2017; 50:353-360. [PMID: 28753520 DOI: 10.1016/j.intimp.2017.07.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/28/2017] [Accepted: 07/19/2017] [Indexed: 01/15/2023]
Abstract
Neuroinflammation plays an important role in the pathogenesis of Parkinson's disease. Interleukin (IL)-10 is one of the most important and best anti-inflammatory cytokines. The objective of this report is to investigate whether IL-10 has any role in protecting ventral mesencephalic (VM) neurons in in vitro model of neuroinflammation. In this study, primary neuron-enriched culture was prepared from the VM tissues of E14 embryos of rats. The cells were pretreated with IL-10 (15 or 50ng/mL) for 1h followed by lipopolysaccharide (LPS, 50ng/mL) application. We found LPS induced neuronal apoptosis and loss while pretreatment with IL-10 reduced neuronal damage after exposure of LPS toxicity. Furthermore, signal transduction pathways related to IL-10 in VM neurons were studied in inflammatory condition. We used both shRNA and pharmacologic inhibition to determine the role of the IL-10 receptor (IL-10R) and its downstream signaling pathways in LPS-induced VM neuronal toxicity. Silence of the IL-10R gene in VM neurons abolished IL-10 mediated protection and the properties of anti-inflammatory and anti-apoptosis. IL-10 also induced phosphorylation of signal transducer and activator of transcription (STAT) 3 in VM neurons. Pretreatment with the specific Janus kinase (JAK) inhibitor reduced STAT3 phosphorylation and blocked IL-10 mediated protection against LPS. These findings suggest that IL-10 provides neuroprotection by acting via IL-10R and its down-stream JAK-STAT3 signal pathways in VM neurons.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
28
|
Ciesielska A, Samaranch L, San Sebastian W, Dickson DW, Goldman S, Forsayeth J, Bankiewicz KS. Depletion of AADC activity in caudate nucleus and putamen of Parkinson's disease patients; implications for ongoing AAV2-AADC gene therapy trial. PLoS One 2017; 12:e0169965. [PMID: 28166239 PMCID: PMC5293261 DOI: 10.1371/journal.pone.0169965] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 12/27/2016] [Indexed: 01/03/2023] Open
Abstract
In Parkinson’s disease (PD), aromatic L-amino acid decarboxylase (AADC) is the rate-limiting enzyme in the conversion of L-DOPA (Sinemet) to dopamine (DA). Previous studies in PD animal models demonstrated that lesion of dopaminergic neurons is associated with profound loss of AADC activity in the striatum, blocking efficient conversion of L-DOPA to DA. Relatively few studies have directly analyzed AADC in PD brains. Thus, the aim of this study was to gain a better understanding of regional changes in AADC activity, DA, serotonin and their monoamine metabolites in the striatum of PD patients and experimentally lesioned animals (rat and MPTP-treated nonhuman primate, NHP). Striatal AADC activity was determined post mortem in neuropathologically confirmed PD subjects, animal models and controls. A regional analysis was performed for striatal AADC activity and monoamine levels in NHP tissue. Interestingly, analysis of putaminal AADC activity revealed that control human striatum contained much less AADC activity than rat and NHP striata. Moreover, a dramatic loss of AADC activity in PD striatum compared to controls was detected. In MPTP-treated NHP, caudate nucleus was almost as greatly affected as putamen, although mean DA turnover was higher in caudate nucleus. Similarly, DA and DA metabolites were dramatically reduced in different regions of PD brains, including caudate nucleus, whereas serotonin was relatively spared. After L-DOPA administration in MPTP-treated NHP, very poor conversion to DA was detected, suggesting that AADC in NHP nigrostriatal fibers is mainly responsible for L-DOPA to DA conversion. These data support further the rationale behind viral gene therapy with AAV2-hAADC to restore AADC levels in putamen and suggest further the advisability of expanding vector delivery to include coverage of anterior putamen and the caudate nucleus.
Collapse
Affiliation(s)
- Agnieszka Ciesielska
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Lluis Samaranch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Waldy San Sebastian
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | | | - Samuel Goldman
- Department of Neurology, University of California San Francisco, San Francisco, CA, United States of America
| | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Krystof S. Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States of America
- * E-mail:
| |
Collapse
|
29
|
Transient glutathione depletion in the substantia nigra compacta is associated with neuroinflammation in rats. Neuroscience 2016; 335:207-20. [DOI: 10.1016/j.neuroscience.2016.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 01/21/2023]
|
30
|
Characterization of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, in rat partial and full nigral 6-hydroxydopamine lesion models of Parkinson's disease. Brain Res 2016; 1646:354-365. [DOI: 10.1016/j.brainres.2016.05.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/20/2022]
|
31
|
Schwerk A, Altschüler J, Roch M, Gossen M, Winter C, Berg J, Kurtz A, Akyüz L, Steiner B. Adipose-derived human mesenchymal stem cells induce long-term neurogenic and anti-inflammatory effects and improve cognitive but not motor performance in a rat model of Parkinson's disease. Regen Med 2016; 10:431-46. [PMID: 26022763 DOI: 10.2217/rme.15.17] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) are easily harvested, and possess anti-inflammatory and trophic properties. Furthermore, MSC promote neuroprotection and neurogenesis, which could greatly benefit neurodegenerative disorders, such as Parkinson's disease. METHODS MSC were transplanted one week after 6-hydroxydopamine lesioning and effects were evaluated after 6 months. RESULTS MSC localized around the substantia nigra and the arachnoid mater, expressing pericyte and endothelial markers. MSC protected dopamine levels and upregulated peripheral anti-inflammatory cytokines. Furthermore, adipose-derived MSC increased neurogenesis in hippocampal and subventricular regions, and boosted memory functioning. CONCLUSION Considering that hyposmia and loss of memory function are two major nonmotor symptoms in Parkinson's disease, transplants with modulatory effects on the hippocampus and subventricular zone could provide a disease-modifying therapy.
Collapse
Affiliation(s)
- Anne Schwerk
- 1Department of Neurology, Charité University Medicine, Berlin, Germany
| | | | - Manfred Roch
- 2Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Manfred Gossen
- 2Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany.,3Helmholtz-Zentrum Geesthacht (HZG), Institute of Biomaterial Science, Teltow, Germany
| | - Christine Winter
- 4Department of Psychiatry, Technical University Dresden, Germany
| | - Jürgen Berg
- 1Department of Neurology, Charité University Medicine, Berlin, Germany
| | - Andreas Kurtz
- 2Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany.,3Helmholtz-Zentrum Geesthacht (HZG), Institute of Biomaterial Science, Teltow, Germany
| | - Levent Akyüz
- 2Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany.,6Institute for Medical Immunology, Berlin, Germany
| | - Barbara Steiner
- 1Department of Neurology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
32
|
Zhu Y, Chen X, Liu Z, Peng YP, Qiu YH. Interleukin-10 Protection against Lipopolysaccharide-Induced Neuro-Inflammation and Neurotoxicity in Ventral Mesencephalic Cultures. Int J Mol Sci 2015; 17:ijms17010025. [PMID: 26729090 PMCID: PMC4730272 DOI: 10.3390/ijms17010025] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/13/2015] [Accepted: 12/18/2015] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL)-10, an anti-inflammatory cytokine, is expressed in the brain and can inhibit microglial activation. Herein, we utilized lipopolysaccharide (LPS)-induced inflammatory Parkinson’s disease (PD) cell model to determine whether microglia and astrocytes are necessary targets for IL-10 neuroprotection. Primary ventral mesencephalic (VM) cultures with different composition of neurons, microglia and astrocytes were prepared. The cells were exposed to IL-10 (15, 50 or 150 ng/mL) 1 h prior to LPS (50 ng/mL) treatment. LPS induced dopaminergic and non-dopaminergic neuronal loss in VM cultures, VM neuron-enriched cultures, and neuron-microglia co-cultures, but not in neuron-astrocyte co-cultures. IL-10 reduced LPS-induced neuronal loss particularly in single VM neuron cultures. Pro-inflammatory mediators (TNF-α, IL-1β, inducible nitric oxide synthase and cyclooxygenase-2) were upregulated in both neuron-microglia and neuron-astrocyte co-cultures by LPS. In contrast, neurotrophic factors (brain-derived neurotrophic factor, insulin-like growth factor-1 or glial cell-derived neurotrophic factor) were downregulated in neuron-microglia co-cultures, but upregulated in neuron-astrocyte co-cultures by LPS. IL-10 reduced both the increase in production of the pro-inflammatory mediators and the decrease in production of the neurotrophic factors induced by LPS. These results suggest that astrocytes can balance LPS neurotoxicity by releasing more neurotrophic factors and that IL-10 exerts neuroprotective property by an extensive action including direct on neurons and indirect via inhibiting microglial activation.
Collapse
Affiliation(s)
- Yan Zhu
- School of Biological & Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China.
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Xiao Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| |
Collapse
|
33
|
Chen T, Hou R, Xu S, Wu C. Donepezil Regulates 1-Methyl-4-phenylpyridinium-Induced Microglial Polarization in Parkinson's Disease. ACS Chem Neurosci 2015; 6:1708-14. [PMID: 26114860 DOI: 10.1021/acschemneuro.5b00026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
1-Methyl-4-phenylpyridinium (MPP+) induces microglial activation and degeneration of dopaminergic (DAergic) neurons. Donepezil is a well-known acetylcholinesterase inhibitor used clinically to treat cognitive dysfunction in Alzheimer's disease (AD). In the present study, we tested the hypothesis that MPP+ promotes microglial M1 polarization and suppresses M2 polarization and that this can be restored by donepezil. Results indicate that MPP+ treatment in microglial BV2 cells promotes microglial polarization toward the M1 state. However, pretreatment with donepezil inhibited MPP+-induced M1 polarization in microglia by suppressing the release of interleukin (IL)-6, IL-1β, or tumor necrosis factor (TNF)-α. Importantly, we found that MPP+ inhibited microglial M2 polarization by suppressing expression of Arg-1, Fizz1, and Ym1, which was also rescued by pretreatment with donepezil. In addition, IL-4-mediated induction of anti-inflammatory marker genes IL-10, IL-13, and transforming growth factor-β2 (TGF-β2) were significantly attenuated by MPP+ in BV2 cells, which was restored by pretreatment with donepezil in a concentration-dependent manner. Mechanistically, we found that the addition of MPP+ reduced the intensity of phosphorylated signal transducer and activator of transcription 6 (STAT6) but not total STAT6 in IL-4-stimulated BV2 cells. Importantly, pretreatment of microglial BV2 cells with donepezil 3 h prior to administration of MPP+ rescued the reduction of STAT6 phosphorylation induced by MPP+.
Collapse
Affiliation(s)
- Teng Chen
- Department
of Neurosurgery, Qilu Hospital of Shandong University, Ji’nan 250012, China
| | - Ruihua Hou
- Clinical
and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO14 3DT, U.K
| | - Shujun Xu
- Department
of Neurosurgery, Qilu Hospital of Shandong University, Ji’nan 250012, China
| | - Chengyuan Wu
- Department
of Neurosurgery, Qilu Hospital of Shandong University, Ji’nan 250012, China
| |
Collapse
|
34
|
Protective Mechanisms of Flavonoids in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:314560. [PMID: 26576219 PMCID: PMC4630416 DOI: 10.1155/2015/314560] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/29/2015] [Indexed: 12/11/2022]
Abstract
Parkinson's disease is a chronic, debilitating neurodegenerative movement disorder characterized by progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta region in human midbrain. To date, oxidative stress is the well accepted concept in the etiology and progression of Parkinson's disease. Hence, the therapeutic agent is targeted against suppressing and alleviating the oxidative stress-induced cellular damage. Within the past decades, an explosion of research discoveries has reported on the protective mechanisms of flavonoids, which are plant-based polyphenols, in the treatment of neurodegenerative disease using both in vitro and in vivo models. In this paper, we have reviewed the literature on the neuroprotective mechanisms of flavonoids in protecting the dopaminergic neurons hence reducing the symptoms of this movement disorder. The mechanism reviewed includes effect of flavonoids in activation of endogenous antioxidant enzymes, suppressing the lipid peroxidation, inhibition of inflammatory mediators, flavonoids as a mitochondrial target therapy, and modulation of gene expression in neuronal cells.
Collapse
|
35
|
Physical Training Regulates Mitochondrial Parameters and Neuroinflammatory Mechanisms in an Experimental Model of Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:261809. [PMID: 26448816 PMCID: PMC4581546 DOI: 10.1155/2015/261809] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/16/2015] [Accepted: 08/06/2015] [Indexed: 11/17/2022]
Abstract
This study aimed to evaluate the effects of two different protocols for physical exercise (strength and aerobic training) on mitochondrial and inflammatory parameters in the 6-OHDA experimental model of Parkinson's disease. Six experimental groups were used (n = 12 per group): untrained + vehicle (Sham), strength training + vehicle (STR), treadmill training + vehicle (TTR), untrained + 6-OHDA (U + 6-OHDA), strength training + 6-OHDA (STR + 6-OHDA), and treadmill training + 6-OHDA (TTR + 6-OHDA). The mice were subjected to strength or treadmill training for 8 weeks. PD was induced via striatal injection of 6-OHDA 24 h after the last exercise session. Mice were euthanized by cervical dislocation and the striatum and hippocampus were homogenized to determine levels of tyrosine hydroxylase (TH), nuclear factor kappa B (NF-κB) p65, and sirtuin 1 (Sirt1) by western blot; tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-17, interferon-γ (IFN-γ), and transforming growth factor β1 (TGF-β1) levels by ELISA; NO content; and complex I (CI) activity. STR + 6-OHDA mice had higher TH levels and CI activity and lower NF-κB p65 and IFN-γ levels in the striatum compared to U + 6-OHDA mice, while TTR + 6-OHDA mice had higher Sirt1 levels and CI activity in both the striatum and the hippocampus, compared to U + 6-OHDA mice. Strength training increased CI activity and TH and Sirt1 levels and reduced NO, NF-κB p65, TNF-α, IFN-γ, IL-1β, and TGF-β1 levels in 6-OHDA mice, while treadmill exercise increased CI activity and NO, TH, and Sirt1 levels and reduced NF-κB p65, TNF-α, IFN-γ, and IL-1β levels. Our results demonstrated that both treadmill training and strength training promote neuroprotection, possibly by stimulating Sirt1 activity, which may in turn regulate both mitochondrial function and neuroinflammation via deacetylation of NF-κB p65. Changes in nitric oxide levels may also be a mechanism by which 6-OHDA-induced inflammation is controlled.
Collapse
|
36
|
Patil RH, Babu RL, Naveen Kumar M, Kiran Kumar KM, Hegde SM, Nagesh R, Ramesh GT, Sharma SC. Anti-Inflammatory Effect of Apigenin on LPS-Induced Pro-Inflammatory Mediators and AP-1 Factors in Human Lung Epithelial Cells. Inflammation 2015; 39:138-147. [DOI: 10.1007/s10753-015-0232-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
Vivekanantham S, Shah S, Dewji R, Dewji A, Khatri C, Ologunde R. Neuroinflammation in Parkinson's disease: role in neurodegeneration and tissue repair. Int J Neurosci 2015; 125:717-25. [PMID: 25364880 DOI: 10.3109/00207454.2014.982795] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Neuroinflammation in Parkinson's disease [PD] is a process that occurs alongside the loss of dopaminergic neurons, and is associated with alterations to many cell types, most notably microglia. This review examines the key evidence contributing to our understanding of the role of inflammation-mediated degeneration of the dopaminergic (DA) nigrostriatal pathway in PD. It will consider the potential role inflammation plays in tissue repair within the brain, inflammation linked gene products that are associated with sporadic Parkinsonian phenotypes (alpha-synuclein, Parkin and Nurr 1), and developing anti-inflammatory drug treatments in PD. With growing evidence supporting the key role of neuroinflammation in PD pathogenesis, new molecular targets are being found that could potentially prevent or delay nigrostriatal DA neuron loss. Hence, this creates the opportunity for disease modifying treatment, to currently what is an incurable disease.
Collapse
Affiliation(s)
- Sayinthen Vivekanantham
- a Imperial College School of Medicine, Imperial College London, Exhibition Road, London , United Kingdom
| | | | | | | | | | | |
Collapse
|
38
|
De Jesús-Cortés H, Miller AD, Britt JK, DeMarco AJ, De Jesús-Cortés M, Stuebing E, Naidoo J, Vázquez-Rosa E, Morlock L, Williams NS, Ready JM, Narayanan NS, Pieper AA. Protective efficacy of P7C3-S243 in the 6-hydroxydopamine model of Parkinson's disease. NPJ PARKINSONS DISEASE 2015; 1. [PMID: 27158662 PMCID: PMC4859442 DOI: 10.1038/npjparkd.2015.10] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background: There are currently no therapeutic options for patients with Parkinson's disease that prevent or slow the death of dopaminergic neurons. We have recently identified the novel P7C3 class of neuroprotective molecules that blocks neuron cell death. AIMS: The aim of this study was to determine whether treatment with highly active members of the P7C3 series blocks dopaminergic neuron cell death and associated behavioral and neurochemical deficits in the rat 6-hydroxydopamine (6-OHDA) model of Parkinson's disease. Methods: After unilateral injection of 6-OHDA into the median forebrain bundle, rats were assessed for behavioral function in the open field, cylinder test, and amphetamine-induced circling test. Thereafter, their brains were subjected to neurochemical and immunohistochemical analysis of dopaminergic neuron survival. Analysis was conducted as a function of treatment with P7C3 compounds, with administration initiated either before or after 6-OHDA exposure. Results: Animals administered P7C3-A20 or P7C3-S243, two of the most advanced agents in the P7C3 series of neuroprotective compounds, both before and after 6-OHDA exposure showed evidence of protective efficacy in all measures. When P7C3-S243 administration was initiated after 6-OHDA exposure, rats also showed protective efficacy in all measures, which included blocking dopaminergic neuron cell death in ipsilateral substantia nigra pars compacta, preservation of dopamine and its metabolites in ipsilateral striatum, and preservation of normal motor behavior. Conclusions: The P7C3 series of compounds may form the basis for developing new therapeutic agents for slowing or preventing progression of Parkinson's disease.
Collapse
Affiliation(s)
- Héctor De Jesús-Cortés
- Graduate program of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Adam D Miller
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jeremiah K Britt
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Anthony J DeMarco
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | - Emily Stuebing
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jacinth Naidoo
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Free Radical and Radiation Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Lorraine Morlock
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nandakumar S Narayanan
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Andrew A Pieper
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Free Radical and Radiation Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; The Iowa City Department of Veterans Affairs, Iowa City, IA, USA
| |
Collapse
|
39
|
Ciesielska A, Sharma N, Beyer J, Forsayeth J, Bankiewicz K. Carbidopa-based modulation of the functional effect of the AAV2-hAADC gene therapy in 6-OHDA lesioned rats. PLoS One 2015; 10:e0122708. [PMID: 25860990 PMCID: PMC4393141 DOI: 10.1371/journal.pone.0122708] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/12/2015] [Indexed: 11/18/2022] Open
Abstract
Progressively blunted response to L-DOPA in Parkinson’s disease (PD) is a critical factor that complicates long-term pharmacotherapy in view of the central importance of this drug in management of the PD-related motor disturbance. This phenomenon is likely due to progressive loss of one of the key enzymes involved in the biosynthetic pathway for dopamine in the basal ganglia: aromatic L-amino acid decarboxylase (AADC). We have developed a gene therapy based on an adeno-associated virus encoding human AADC (AAV2-hAADC) infused into the Parkinsonian striatum. Although no adverse clinical effects of the AAV2-hAADC gene therapy have been observed so far, the ability to more precisely regulate transgene expression or transgene product activity could be an important long-term safety feature. The present study was designed to define pharmacological regulation of the functional activity of AAV2-hAADC transgene product by manipulating L-DOPA and carbidopa (AADC inhibitor) administration in hemi-parkinsonian rats. Thirty days after unilateral striatal infusion of AAV2-hAADC, animals displayed circling behavior and acceleration of dopamine metabolism in the lesioned striatum after administration of a low dose of L-DOPA (5 mg/kg) co-administered with 1.25 mg/kg of carbidopa. This phenomenon was not observed in control AAV2-GFP-treated rats. Withdrawal of carbidopa from a daily L-DOPA regimen decreased the peripheral L-DOPA pool, resulting in almost total loss of L-DOPA-induced behavioral response in AAV2-hAADC rats and a significant decline in striatal dopamine turnover. The serum L-DOPA level correlated with the magnitude of circling behavior in AAV2-hAADC rats. Additionally, AADC activity in homogenates of lesioned striata transduced by AAV2-AADC was 10-fold higher when compared with AAV2-GFP-treated control striata, confirming functional transduction. Our data suggests that the pharmacological regulation of circulating L-DOPA might be effective in the controlling of function of AAV2-hAADC transgene product in PD gene therapy.
Collapse
Affiliation(s)
- Agnieszka Ciesielska
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United State of America
| | - Nitasha Sharma
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United State of America
| | - Janine Beyer
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United State of America
| | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United State of America
| | - Krystof Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United State of America
- * E-mail:
| |
Collapse
|
40
|
Cebrián C, Loike JD, Sulzer D. Neuroinflammation in Parkinson's disease animal models: a cell stress response or a step in neurodegeneration? Curr Top Behav Neurosci 2015; 22:237-270. [PMID: 25293443 DOI: 10.1007/7854_2014_356] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The motor symptoms of Parkinson's disease are due to the progressive degeneration of dopaminergic neurons in the substantia nigra. Multiple neuroinflammatory processes are exacerbated in Parkinson's disease, including glial-mediated reactions, increased expression of proinflammatory substances, and lymphocytic infiltration, particularly in the substantia nigra. Neuroinflammation is also implicated in the neurodegeneration and consequent behavioral symptoms of many Parkinson's disease animal models, although it is not clear whether these features emulate pathogenic steps in the genuine disorder or if some inflammatory features provide protective stress responses. Here, we compare and summarize findings on neuroinflammatory responses and effects on behavior in a wide range of toxin-based, inflammatory and genetic Parkinson's disease animal models.
Collapse
Affiliation(s)
- Carolina Cebrián
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | | | | |
Collapse
|
41
|
Pisanu A, Lecca D, Mulas G, Wardas J, Simbula G, Spiga S, Carta AR. Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson's disease. Neurobiol Dis 2014; 71:280-91. [DOI: 10.1016/j.nbd.2014.08.011] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 11/25/2022] Open
|
42
|
Joniec-Maciejak I, Ciesielska A, Wawer A, Sznejder-Pachołek A, Schwenkgrub J, Cudna A, Hadaczek P, Bankiewicz KS, Członkowska A, Członkowski A. The influence of AAV2-mediated gene transfer of human IL-10 on neurodegeneration and immune response in a murine model of Parkinson's disease. Pharmacol Rep 2014; 66:660-9. [PMID: 24948069 DOI: 10.1016/j.pharep.2014.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 03/09/2014] [Accepted: 03/13/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND The aim of this study was to examine the effect of AAV2-hIL-10 (vector containing cDNA for human interleukin 10) on dopaminergic system activity (measured as DA levels and TH mRNA expression in mouse striata), and other monoamine and amino acid neurotransmitters concentration as well as development of inflammatory processes (measured as TGF-β, IFN-γ and GFAP mRNA expression) in a murine MPTP neurotoxicant model of Parkinson's disease. METHODS Male C57BL/6 mice 12 months-old were used in this study. AAV2-hIL-10 vector was bilaterally administered into striatum at 14, 21 or 28 days prior to MPTP intoxication. Animals were sacrificed at 7 days following MPTP injection. The expression of hIL-10 (human interleukin 10) was examined by ELISA. Striatal monoamine and amino acid neurotransmitters were measured by HPLC method. TH, TGF-β, IFN-γ and GFAP mRNA expression was examined by RT-PCR method. RESULTS MPTP treatment dramatically reduced DA levels and decreased TH mRNA expression in mouse striata, effects that were significantly impeded by AAV2-hIL-10 administration prior to MPTP intoxication. AAV2-hIL-10 infusion increased IFN-γ, TGF-β and GFAP mRNA expression. CONCLUSIONS Our data suggest that the transfer of AAV2-hIL-10 into the striatum may play a neuroprotective role in the mouse MPTP model of PD and these effects are mediated by the anti-inflammatory action of IL-10.
Collapse
Affiliation(s)
- Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland.
| | - Agnieszka Ciesielska
- Department of Neurosurgery, University of California at San Francisco, San Francisco, CA, USA
| | - Adriana Wawer
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Anna Sznejder-Pachołek
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Schwenkgrub
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudna
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Piotr Hadaczek
- Department of Neurosurgery, University of California at San Francisco, San Francisco, CA, USA
| | - Kristof S Bankiewicz
- Department of Neurosurgery, University of California at San Francisco, San Francisco, CA, USA
| | - Anna Członkowska
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Andrzej Członkowski
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
43
|
Wheeler CJ, Seksenyan A, Koronyo Y, Rentsendorj A, Sarayba D, Wu H, Gragg A, Siegel E, Thomas D, Espinosa A, Thompson K, Black K, Koronyo-Hamaoui M, Pechnick R, Irvin DK. T-Lymphocyte Deficiency Exacerbates Behavioral Deficits in the 6-OHDA Unilateral Lesion Rat Model for Parkinson's Disease. ACTA ACUST UNITED AC 2014; 5. [PMID: 25346865 PMCID: PMC4207300 DOI: 10.4172/2155-9562.1000209] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
T-lymphocytes have been previously implicated in protecting dopaminergic neurons in the substantianigra from induced cell death. However, the role of T-cells in neurodegenerative models such as Parkinson’s disease (PD) has not been fully elucidated. To examine the role of T-lymphocytes on motor behavior in the 6-hydroxydopamine (6-OHDA) unilateral striatal partial lesion PD rat model, we assessed progression of hemi-parkinsonian lesions in the substantia nigra, induced by 6-OHDA striatal injections, in athymic rats (RNU−/−, T-lymphocyte-deficient) as compared to RNU−/+ rats (phenotypically normal). Motor skills were determined by the cylinder and D-amphetamine sulfate-induced rotational behavioral tests. Cylinder behavioral test showed no significant difference between unilaterally lesioned RNU−/− and RNU−/+ rats. However both unilaterally lesioned RNU−/− and RNU−/+ rats favored the use of the limb ipsilateral to lesion. Additionally, amphetamine-induced rotational test revealed greater rotational asymmetry in RNU−/− rats compared to RNU−/+ rats at two- and six-week post-lesion. Quantitative immunohistochemistry confirmed loss of striatal TH-immunopositive fibers in RNU−/− and RNU−/+ rat, as well as blood-brain-barrier changes associated with PD that may influence passage of immune cells into the central nervous system in RNU−/− brains. Specifically, GFAP immunopositive cells were decreased, as were astrocytic end-feet (AQP4) contacting blood vessels (laminin) in the lesioned relative to contralateral striatum. Flow cytometric analysis in 6-OHDA lesioned RNU−/+rats revealed increased CD4+ and decreased CD8+ T cells specifically within lesioned brain. These results suggest that both major T cell subpopulations are significantly and reciprocally altered following 6-OHDA-lesioning, and that global T cell deficiency exacerbates motor behavioral defects in this rat model of PD.
Collapse
Affiliation(s)
- Christopher J Wheeler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Akop Seksenyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Danielle Sarayba
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Henry Wu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Ashley Gragg
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Emily Siegel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Deborah Thomas
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Andres Espinosa
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Kerry Thompson
- Department of Biology, Occidental College, Los Angeles, CA 90041, USA
| | - Keith Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| | - Robert Pechnick
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, CA 91766, USA
| | - Dwain K Irvin
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA, Department of Psychiatry, Cedars-Sinai Medical Center, Los Angeles, California, 90048, USA; Occidental College, Los Angeles, CA 90041, USA
| |
Collapse
|
44
|
Effect of human interleukin-10 on the expression of nitric oxide synthases in the MPTP-based model of Parkinson's disease. Pharmacol Rep 2013; 65:44-9. [PMID: 23563022 DOI: 10.1016/s1734-1140(13)70962-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 09/27/2012] [Indexed: 01/28/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder. The degeneration of the nigro-striatal pathway has been linked with the inflammatory process accompanied by the robust up-regulation of the nitric oxide synthase (NOS) and production of the neurotoxic level of nitric oxide (NO). One of the therapeutic strategies of PD is based on the reduction of the detrimental neuroinflammatory markers in the lesioned nigro-striatal pathway. In this study we have investigated the neuroprotective effect of the cerebral infusion of recombinant adeno-associated viral vector, expressing the gene for human interleukin-10 (AAV2-hIL-10) in a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). It is known that IL-10 is a potent anti-inflammatory cytokine that limits the inducible nitric oxide synthase (iNOS) gene expression. METHODS The striatal iNOS, neuronal nitric oxide synthase (nNOS) and tyrosine hydroxylase (TH) protein expression was evaluated by immunoblot analysis. RESULTS The intracerebral injection of the AAV2-hIL-10, before the lesion, induced the upregulation of the striatal TH protein, depleted by MPTP intoxication. This AAV2-hIL-10-induced increase of TH level was associated with the suppression of iNOS protein expression in the lesioned striatum. CONCLUSION The results revealed protective properties of AAV2-hIL-10.
Collapse
|
45
|
Cappellano G, Carecchio M, Fleetwood T, Magistrelli L, Cantello R, Dianzani U, Comi C. Immunity and inflammation in neurodegenerative diseases. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:89-107. [PMID: 23844334 PMCID: PMC3703122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 06/11/2013] [Indexed: 06/02/2023]
Abstract
Immune reactions inside the central nervous system are finely regulated, thanks to the presence of several checkpoints that have the fundamental purpose to preserve this fragile tissue form harmful events. The current knowledge on the role of neuroinflammation and neuro-immune interactions in the fields of multiple sclerosis, Alzheimer's disease and Parkinson's disease is reviewed. Moreover, a focus on the potential role of both active and passive immunotherapy is provided. Finally, we propose a common perspective, which implies that, under pathological conditions, inflammation may exert both detrimental and protective functions, depending on local factors and the timing of immune activation and shutting-off systems.
Collapse
Affiliation(s)
- Giuseppe Cappellano
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Health Sciences, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Miryam Carecchio
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Thomas Fleetwood
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Luca Magistrelli
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Roberto Cantello
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Health Sciences, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Cristoforo Comi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| |
Collapse
|
46
|
vinh quôc Luong K, Thi Hoàng Nguyên L. Vitamin D and Parkinson's disease. J Neurosci Res 2012; 90:2227-36. [DOI: 10.1002/jnr.23115] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 06/21/2012] [Indexed: 01/11/2023]
|
47
|
Modulating Microglia Activity with PPAR-γ Agonists: A Promising Therapy for Parkinson’s Disease? Neurotox Res 2012; 23:112-23. [DOI: 10.1007/s12640-012-9342-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/08/2012] [Accepted: 07/18/2012] [Indexed: 12/11/2022]
|
48
|
Miranpuri GS, Kumbier L, Hinchman A, Schomberg D, Wang A, Marshall H, Kubota K, Ross C, Sillay K. Gene-based therapy of Parkinson's Disease: Translation from animal model to human clinical trial employing convection enhanced delivery. Ann Neurosci 2012; 19:133-46. [PMID: 25205986 PMCID: PMC4117084 DOI: 10.5214/ans.0972.7531.190310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 07/02/2012] [Accepted: 07/02/2012] [Indexed: 11/17/2022] Open
Abstract
The existing treatment of Parkinson's disease (PD) is directed towards substituting dopamine loss with either dopamine replacement therapy or pharmacological therapies aimed at increasing dopamine at the synapse level. Emerging viable alternatives include the use of cell-based and gene-based therapeutics. In this review, we discuss efforts in developing in vitro and in vivo models and their translation to human clinical trials for gene-based therapy of this distressing and prevalent neurodegenerative disorder. Given the mismatch between expectations from preclinical data and results of human pivotal trials, drug delivery has been identified as the key emerging area for translational research due to limitation of limited efficacy. The chief highlights of the current topic include use of improved delivery methods of gene-based therapeutic agents. Convection-enhanced delivery (CED), an advanced infusion technique with demonstrated utility in ex vivo and in vivo animal models has recently been adopted for PD gene-based therapy trials. Several preclinical studies suggest that magnetic resonance imaging (MRI)-guided navigation for accurately targeting and real time monitoring viral vector delivery (rCED) in future clinical trials involving detection of gene expression and restoration of dopaminergic function loss using pro-drug approach will greatly enhance these PD treatments.
Collapse
Affiliation(s)
- Gurwattan S. Miranpuri
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Lauren Kumbier
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Angelica Hinchman
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Dominic Schomberg
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Anyi Wang
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Hope Marshall
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Ken Kubota
- Kinetic Foundation, Los Altos, CA, 94023, USA
| | - Chris Ross
- Engineering Resources Group Inc, Pembroke Pines, FL, 33029, USA
| | - Karl Sillay
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| |
Collapse
|
49
|
López-Villodres JA, De La Cruz JP, Muñoz-Marin J, Guerrero A, Reyes JJ, González-Correa JA. Cytoprotective effect of nonsteroidal antiinflammatory drugs in rat brain slices subjected to reoxygenation after oxygen–glucose deprivation. Eur J Pharm Sci 2012; 45:624-31. [DOI: 10.1016/j.ejps.2012.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 12/15/2011] [Accepted: 01/02/2012] [Indexed: 11/15/2022]
|
50
|
Peterson LJ, Flood PM. Oxidative stress and microglial cells in Parkinson's disease. Mediators Inflamm 2012; 2012:401264. [PMID: 22544998 PMCID: PMC3321615 DOI: 10.1155/2012/401264] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 12/22/2022] Open
Abstract
Significant evidence has now been accumulated that microglial cells play a central role in the degeneration of DA neurons in animal models of PD. The oxidative stress response by microglial cells, most notably the activity of the enzyme NADPH oxidase, appears to play a central role in the pathology of PD. This oxidative stress response occurs in microglia through the activation of the ERK signaling pathway by proinflammatory stimuli, leading to the phosphorylation and translocation of the p47(phox) and p67(phox) cytosolic subunits, the activation of membrane-bound PHOX, and the production of ROS. Therapeutic anti-inflammatories which prevent DA neurodegeneration in PD, including anti-inflammatory cytokines, morphinan compounds, NADPH oxidase inhibitors, NF-κB inhibitors, and β2-AR agonists, all function to inhibit the activation of the PHOX in microglial cells. These observations suggest a central role for the oxidative stress response in microglial cells as a mediator or regulator of DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Lynda J. Peterson
- North Carolina Oral Health Institute, The University of North Carolina at Chapel Hill, CB#7454, Chapel Hill, NC 27599-7454, USA
| | - Patrick M. Flood
- North Carolina Oral Health Institute, The University of North Carolina at Chapel Hill, CB#7454, Chapel Hill, NC 27599-7454, USA
| |
Collapse
|