51
|
Chen Z, Chen S, Liu J. The role of T cells in the pathogenesis of Parkinson's disease. Prog Neurobiol 2018; 169:1-23. [PMID: 30114440 DOI: 10.1016/j.pneurobio.2018.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/24/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023]
Abstract
Recent evidence has shown that neuroinflammation plays a key role in the pathogenesis of Parkinson's disease (PD). However, different components of the brain's immune system may exert diverse effects on neuroinflammatory events in PD. The adaptive immune response, especially the T cell response, can trigger type 1 pro-inflammatory activities and suppress type 2 anti-inflammatory activities, eventually resulting in deregulated neuroinflammation and subsequent dopaminergic neurodegeneration. Additionally, studies have increasingly shown that therapies targeting T cells can alleviate neurodegeneration and motor behavior impairment in animal models of PD. Therefore, we conclude that abnormal T cell-mediated immunity is a fundamental pathological process that may be a promising translational therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhichun Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
52
|
Ferreira SA, Romero-Ramos M. Microglia Response During Parkinson's Disease: Alpha-Synuclein Intervention. Front Cell Neurosci 2018; 12:247. [PMID: 30127724 PMCID: PMC6087878 DOI: 10.3389/fncel.2018.00247] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
The discovery of the central role played by the protein alpha-synuclein in Parkinson's disease and other Lewy body brain disorders has had a great relevance in the understanding of the degenerative process occurring in these diseases. In addition, during the last two decades, the evidence suggesting an immune response in Parkinson's disease patients have multiplied. The role of the immune system in the disease is supported by data from genetic studies and patients, as well as from laboratory animal models and cell cultures. In the immune response, the microglia, the immune cell of the brain, will have a determinant role. Interestingly, alpha-synuclein is suggested to have a central function not only in the neuronal events occurring in Parkinson's disease, but also in the immune response during the disease. Numerous studies have shown that alpha-synuclein can act directly on immune cells, such as microglia in brain, initiating a sterile response that will have consequences for the neuronal health and that could also translate in a peripheral immune response. In parallel, microglia should also act clearing alpha-synuclein thus avoiding an overabundance of the protein, which is crucial to the disease progression. Therefore, the microglia response in each moment will have significant consequences for the neuronal fate. Here we will review the literature addressing the microglia response in Parkinson's disease with an especial focus on the protein alpha-synuclein. We will also reflect upon the limitations of the studies carried so far and in the therapeutic possibilities opened based on these recent findings.
Collapse
Affiliation(s)
- Sara A Ferreira
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- AU IDEAS center NEURODIN, Aarhus University, Aarhus, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
53
|
Linking Neuroinflammation and Neurodegeneration in Parkinson's Disease. J Immunol Res 2018; 2018:4784268. [PMID: 29850629 PMCID: PMC5926497 DOI: 10.1155/2018/4784268] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/28/2018] [Indexed: 11/30/2022] Open
Abstract
Neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD) impose a pressing burden on our developed and consequently aging society. Misfolded protein aggregates are a critical aspect of several neurodegenerative diseases. Nevertheless, several questions remain unanswered regarding the role of misfolded protein aggregates and the cause of neuronal cell death. Recently, it has been postulated that neuroinflammatory processes might play a crucial role in the pathogenesis of PD. Numerous postmortem, brain imaging, epidemiological, and animal studies have documented the involvement of the innate and adaptive immunity in neurodegeneration. Whether these inflammatory processes are directly involved in the etiology of PD or represent secondary consequences of nigrostriatal pathway injury is the subject of intensive research. Immune alterations in response to extracellular α-synuclein may play a critical role in modulating Parkinson's disease progression. In this review, we address the current concept of neuroinflammation and its involvement in PD-associated neurodegeneration.
Collapse
|
54
|
Mochizuki H, Choong CJ, Masliah E. A refined concept: α-synuclein dysregulation disease. Neurochem Int 2018; 119:84-96. [PMID: 29305061 DOI: 10.1016/j.neuint.2017.12.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/21/2017] [Accepted: 12/29/2017] [Indexed: 12/23/2022]
Abstract
α-synuclein (αSyn) still remains a mysterious protein even two decades after SNCA encoding it was identified as the first causative gene of familial Parkinson's disease (PD). Accumulation of αSyn causes α-synucleinopathies including PD, dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Recent advances in therapeutic approaches offer new antibody-, vaccine-, antisense-oligonucleotide- and small molecule-based options to reduce αSyn protein levels and aggregates in patient's brain. Gathering research information of other neurological disease particularly Alzheimer's disease, recent disappointment of an experimental amyloid plaques busting antibody in clinical trials underscores the difficulty of treating people who show even mild dementia as damage in their brain may already be too extensive. Prodromal intervention to inhibit the accumulation of pathogenic protein may advantageously provide a better outcome. However, treatment prior to onset is not ethically justified as standard practice at present. In this review, we initiate a refined concept to define early pathogenic state of αSyn accumulation before occurrence of brain damage as a disease criterion for αSyn dysregulation disease.
Collapse
Affiliation(s)
- Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| | - Chi-Jing Choong
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Eliezer Masliah
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
55
|
McGee DJ, Lu XH, Disbrow EA. Stomaching the Possibility of a Pathogenic Role for Helicobacter pylori in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2018; 8:367-374. [PMID: 29966206 PMCID: PMC6130334 DOI: 10.3233/jpd-181327] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Abstract
While a small subset of Parkinson's disease cases have genetic causes, most cases are sporadic and may have an environmental contributor that has largely remained enigmatic. Remarkably, gastrointestinal symptoms in PD patients serve as a prodrome for the eventual motor dysfunctions. Herein, we review studies exploring a possible link between the gastric human pathogen Helicobacter pylori and PD. We provide plausible and testable hypotheses for how this organism might contribute to PD: 1) a toxin(s) produced by the bacteria; 2) disruption of the intestinal microbiome; 3) local inflammation that crosses the gut-brain axis, leading to neuroinflammation; and 4) manipulation of the pharmacokinetics of the PD drug levodopa by H. pylori, even in those not receiving exogenous levodopa. Key findings are: 1) people with PD are 1.5-3-fold more likely to be infected with H. pylori than people without PD; 2) H. pylori-infected PD patients display worse motor functions than H. pylori-negative PD patients; 3) eradication of H. pylori improves motor function in PD patients over PD patients whose H. pylori was not eradicated; and 4) eradication of H. pylori improves levodopa absorption in PD patients compared to that of PD patients whose H. pylori was not eradicated. Evidence is accumulating that H. pylori has a link with PD, but the mechanism is unclear. Future work should explore the effects of H. pylori on development of PD in defined PD animal models, focusing on the roles of H. pylori toxins, inflammation, levodopa absorption, and microbiome dysbiosis.
Collapse
Affiliation(s)
- David J. McGee
- Department of Microbiology and Immunology, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Xiao-Hong Lu
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Elizabeth A. Disbrow
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
- Department of Neurology, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| |
Collapse
|
56
|
Bengoa-Vergniory N, Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Alpha-synuclein oligomers: a new hope. Acta Neuropathol 2017; 134:819-838. [PMID: 28803412 PMCID: PMC5663814 DOI: 10.1007/s00401-017-1755-1] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 01/22/2023]
Abstract
Alpha-synuclein is a protein implicated in Parkinson’s disease and thought to be one of the main pathological drivers in the disease, although it remains unclear how this protein elicits its neurotoxic effects. Recent findings indicate that the assembly of toxic oligomeric species of alpha-synuclein may be one of the key processes for the pathology and spread of the disease. The absence of a sensitive in situ detection method has hindered the study of these oligomeric species and the role they play in the human brain until recently. In this review, we assess the evidence for the toxicity and prion-like activity of oligomeric forms of alpha-synuclein and discuss the advances in our understanding of the role of alpha-synuclein in Parkinson’s disease that may be brought about by the specific and sensitive detection of distinct oligomeric species in post-mortem patient brain. Finally, we discuss current approaches being taken to therapeutically target alpha-synuclein oligomers and their implications.
Collapse
Affiliation(s)
- Nora Bengoa-Vergniory
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford, OX1 3QT, UK
| | - Rosalind F Roberts
- Montreal Neurological Institute, McGill University, 3801 Rue University, Montreal, QC, H3A 2B4, Canada
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford, OX1 3QT, UK.
| | - Javier Alegre-Abarrategui
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
57
|
Villadiego J, Labrador-Garrido A, Franco JM, Leal-Lasarte M, De Genst EJ, Dobson CM, Pozo D, Toledo-Aral JJ, Roodveldt C. Immunization with α-synuclein/Grp94 reshapes peripheral immunity and suppresses microgliosis in a chronic Parkinsonism model. Glia 2017; 66:191-205. [PMID: 29024008 DOI: 10.1002/glia.23237] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022]
Abstract
Neuroinflammation mediated by chronically activated microglia, largely caused by abnormal accumulation of misfolded α-synuclein (αSyn) protein, is known to contribute to the pathophysiology of Parkinson's disease (PD). In this work, based on the immunomodulatory activities displayed by particular heat-shock proteins (HSPs), we tested a novel vaccination strategy that used a combination of αSyn and Grp94 (HSPC4 or Gp96) chaperone and a murine PD model. We used two different procedures, first, the adoptive transfer of splenocytes from αSyn/Grp94-immunized mice to recipient animals, and second, direct immunization with αSyn/Grp94, to study the effects in a chronic mouse MPTP-model of parkinsonism. We found that both approaches promoted a distinct profile in the peripheral system-supported by humoral and cellular immunity-consisting of a Th1-shifted αSyn-specific response accompanied by an immune-regulatory/Th2-skewed general phenotype. Remarkably, this mixed profile sustained by αSyn/Grp94 immunization led to strong suppression of microglial activation in the substantia nigra and striatum, pointing to a newly described positive effect of anti-αSyn Th1-responses in the context of PD. This strategy is the first to target αSyn and report the suppression of PD-associated microgliosis. Overall, we show that the αSyn/Grp94 combination supports a distinct and long-lasting immune profile in the peripheral system, which has an impact at the CNS level by suppressing chronic microglial activation in an MPTP model of PD. Furthermore, our study demonstrates that reshaping peripheral immunity by vaccination with appropriate misfolding protein/HSP combinations could be highly beneficial as a treatment for neurodegenerative misfolding diseases.
Collapse
Affiliation(s)
- Javier Villadiego
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio-CSIC-Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Adahir Labrador-Garrido
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER. Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain.,Departamento de Bioquímica Médica, Biología Molecular e Inmunología. Universidad de Sevilla, Seville, Spain
| | - Jaime M Franco
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER. Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain.,Departamento de Bioquímica Médica, Biología Molecular e Inmunología. Universidad de Sevilla, Seville, Spain
| | - Magdalena Leal-Lasarte
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER. Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - Erwin J De Genst
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | | | - David Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER. Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain.,Departamento de Bioquímica Médica, Biología Molecular e Inmunología. Universidad de Sevilla, Seville, Spain
| | - Juan J Toledo-Aral
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio-CSIC-Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Cintia Roodveldt
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER. Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|
58
|
Dhillon JKS, Riffe C, Moore BD, Ran Y, Chakrabarty P, Golde TE, Giasson BI. A novel panel of α-synuclein antibodies reveal distinctive staining profiles in synucleinopathies. PLoS One 2017; 12:e0184731. [PMID: 28910367 PMCID: PMC5599040 DOI: 10.1371/journal.pone.0184731] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 12/25/2022] Open
Abstract
Synucleinopathies are a spectrum of neurodegenerative diseases characterized by the intracellular deposition of the protein α-synuclein leading to multiple outcomes, including dementia and Parkinsonism. Recent findings support the notion that across the spectrum of synucleinopathies there exist diverse but specific biochemical modifications and/or structural conformations of α-synuclein, which would give rise to protein strain specific prion-like intercellular transmission, a proposed model that could explain synucleinopathies disease progression. Herein, we characterized a panel of antibodies with epitopes within both the C- and N- termini of α-synuclein. A comprehensive analysis of human pathological tissue and mouse models of synucleinopathy with these antibodies support the notion that α-synuclein exists in distinct modified forms and/or structural variants. Furthermore, these well-characterized and specific tools allow the investigation of biochemical changes associated with α-synuclein inclusion formation. We have identified several antibodies of interest with diverse staining and epitope properties that will prove useful in future investigations of strain specific disease progression and the development of targeted immunotherapeutic approaches to synucleinopathies.
Collapse
Affiliation(s)
- Jess-Karan S. Dhillon
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Cara Riffe
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Brenda D. Moore
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Yong Ran
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Todd E. Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Benoit I. Giasson
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
59
|
Inhibition of the JAK/STAT Pathway Protects Against α-Synuclein-Induced Neuroinflammation and Dopaminergic Neurodegeneration. J Neurosci 2017; 36:5144-59. [PMID: 27147665 DOI: 10.1523/jneurosci.4658-15.2016] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/22/2016] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Parkinson's Disease (PD) is an age-related, chronic neurodegenerative disorder. At present, there are no disease-modifying therapies to prevent PD progression. Activated microglia and neuroinflammation are associated with the pathogenesis and progression of PD. Accumulation of α-synuclein (α-SYN) in the brain is a core feature of PD and leads to microglial activation, inflammatory cytokine/chemokine production, and ultimately to neurodegeneration. Given the importance of the JAK/STAT pathway in activating microglia and inducing cytokine/chemokine expression, we investigated the therapeutic potential of inhibiting the JAK/STAT pathway using the JAK1/2 inhibitor, AZD1480. In vitro, α-SYN exposure activated the JAK/STAT pathway in microglia and macrophages, and treatment with AZD1480 inhibited α-SYN-induced major histocompatibility complex Class II and inflammatory gene expression in microglia and macrophages by reducing STAT1 and STAT3 activation. For in vivo studies, we used a rat model of PD induced by viral overexpression of α-SYN. AZD1480 treatment inhibited α-SYN-induced neuroinflammation by suppressing microglial activation, macrophage and CD4(+) T-cell infiltration and production of proinflammatory cytokines/chemokines. Numerous genes involved in cell-cell signaling, nervous system development and function, inflammatory diseases/processes, and neurological diseases are enhanced in the substantia nigra of rats with α-SYN overexpression, and inhibited upon treatment with AZD1480. Importantly, inhibition of the JAK/STAT pathway prevented the degeneration of dopaminergic neurons in vivo These results indicate that inhibiting the JAK/STAT pathway can prevent neuroinflammation and neurodegeneration by suppressing activation of innate and adaptive immune responses to α-SYN. Furthermore, this suggests the feasibility of targeting the JAK/STAT pathway as a neuroprotective therapy for neurodegenerative diseases. SIGNIFICANCE STATEMENT α-SYN plays a central role in the pathophysiology of PD through initiation of neuroinflammatory responses. Using an α-SYN overexpression PD model, we demonstrate a beneficial therapeutic effect of AZD1480, a specific inhibitor of JAK1/2, in suppressing neuroinflammation and neurodegeneration. Our findings document that inhibition of the JAK/STAT pathway influences both innate and adaptive immune responses by suppressing α-SYN-induced microglia and macrophage activation and CD4(+) T-cell recruitment into the CNS, ultimately suppressing neurodegeneration. These findings are the first documentation that suppression of the JAK/STAT pathway disrupts the circuitry of neuroinflammation and neurodegeneration, thus attenuating PD pathogenesis. JAK inhibitors may be a viable therapeutic option for the treatment of PD patients.
Collapse
|
60
|
von Euler Chelpin M, Vorup-Jensen T. Targets and Mechanisms in Prevention of Parkinson's Disease through Immunomodulatory Treatments. Scand J Immunol 2017; 85:321-330. [PMID: 28231624 DOI: 10.1111/sji.12542] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 01/13/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world; however, there is no cure for it. Current treatments only relieve some of the symptoms, without ceasing the disease, and lose efficacy with prolonged treatment. Considerable evidence shows that persistent inflammatory responses, involving T cell infiltration and glial cell activation, are common characteristics of human patients and play a crucial role in the degeneration of dopaminergic neurons. Therefore, it is important to develop therapeutic strategies that can impede or halt the disease through the modulation of the peripheral immune system by aiming at controlling the existing neuroinflammation. Most of the immunomodulatory therapies designed for the treatment of Parkinson's disease are based on vaccines using AS or antibodies against it; yet, it is of significant interest to explore other formulations that could be used as therapeutic agents. Several vaccination procedures have shown that inducing regulatory T cells in the periphery is protective in PD animal models. In this regard, the formulation glatiramer acetate (Copaxone® ), extensively used for the treatment of multiple sclerosis, could be a suitable candidate due to its capability to increase the number and suppressor capacity of regulatory T cells. In this review, we will present some of the recent immunomodulatory therapies for PD including vaccinations with AS or glatiramoids, or both, as treatments of PD pathology.
Collapse
Affiliation(s)
| | - T Vorup-Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
61
|
Involvement of the kynurenine pathway in the pathogenesis of Parkinson’s disease. Prog Neurobiol 2017; 155:76-95. [DOI: 10.1016/j.pneurobio.2015.12.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022]
|
62
|
Arevalo-Villalobos JI, Govea-Alonso DO, Monreal-Escalante E, Zarazúa S, Rosales-Mendoza S. LTB-Syn: a recombinant immunogen for the development of plant-made vaccines against synucleinopathies. PLANTA 2017; 245:1231-1239. [PMID: 28315001 DOI: 10.1007/s00425-017-2675-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/08/2017] [Indexed: 06/06/2023]
Abstract
MAIN CONCLUSION A recombinant antigen targeting α-synuclein was produced in the plant cell rendering an immunogenic protein capable to induce humoral responses in mice upon oral administration. Synucleinopathies are neurodegenerative diseases characterized by the abnormal accumulation of α-synuclein (α-Syn, a 140 amino acid protein that normally plays various neurophysiologic roles) aggregates. Parkinson's disease (PD) is the synucleinopathy with the highest epidemiologic impact and although its etiology remains unknown, α-Syn aggregation during disease progression pointed out α-Syn as target in the development of immunotherapies. Herein a chimeric protein, comprising the B subunit of the enterotoxin from enterotoxigenic Escherichia coli and α-Syn epitopes, was expressed in the plant cell having the potential to induce humoral responses following oral immunization. This approach will serve as the basis for the development of oral plant-based vaccines against PD with several potential advantages such as low cost, easy scale-up during production, and easy administration.
Collapse
Affiliation(s)
- Jaime I Arevalo-Villalobos
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Dania O Govea-Alonso
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Elizabeth Monreal-Escalante
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Sergio Zarazúa
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico.
| |
Collapse
|
63
|
Duffy SS, Keating BA, Perera CJ, Moalem-Taylor G. The role of regulatory T cells in nervous system pathologies. J Neurosci Res 2017; 96:951-968. [DOI: 10.1002/jnr.24073] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/28/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Samuel S. Duffy
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Brooke A. Keating
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Chamini J. Perera
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Gila Moalem-Taylor
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| |
Collapse
|
64
|
Tyson T, Steiner JA, Brundin P. Sorting out release, uptake and processing of alpha-synuclein during prion-like spread of pathology. J Neurochem 2016; 139 Suppl 1:275-289. [PMID: 26617280 PMCID: PMC4958606 DOI: 10.1111/jnc.13449] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022]
Abstract
Parkinson's disease is a progressive neurological disorder that is characterized by the formation of intracellular protein inclusion bodies composed primarily of a misfolded and aggregated form of the protein α-synuclein. There is growing evidence that supports the prion-like hypothesis of α-synuclein progression. This hypothesis postulates that α-synuclein is a prion-like pathological agent and is responsible for the progression of Parkinson pathology in the brain. Potential misfolding or aggregation of α-synuclein that might occur in the peripheral nervous system as a result of some insult, environmental or genetic (or more likely a combination of both) that might spread into the midbrain, eventually causing degeneration of the neurons in the substantia nigra. As the disease progresses further, it is likely that α-synuclein pathology continues to spread throughout the brain, including the cortex, leading to deterioration of cognition and higher brain functions. While it is unknown why α-synuclein initially misfolds and aggregates, a great deal has been learned about how the cell handles aberrant α-synuclein assemblies. In this review, we focus on these mechanisms and discuss them in an attempt to define the role that they might play in the propagation of misfolded α-synuclein from cell-to-cell. The prion-like hypothesis of α-synuclein pathology suggests a method for the transmission of misfolded α-synuclein from one neuron to another. This hypothesis postulates that misfolded α-synuclein becomes aggregation prone and when released and taken up by neighboring cells, seeds further misfolding and aggregation. In this review we examine the cellular mechanisms that are involved in the processing of α-synuclein and how these may contribute to the prion-like propagation of α-synuclein pathology. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
65
|
Choong CJ, Mochizuki H. Gene therapy targeting mitochondrial pathway in Parkinson's disease. J Neural Transm (Vienna) 2016; 124:193-207. [PMID: 27638713 DOI: 10.1007/s00702-016-1616-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) presents a relative selective localization of pathology to substantia nigra and well-defined motor symptoms caused by dopaminergic degeneration that makes it an ideal target for gene therapy. Parallel progress in viral vector systems enables the delivery of therapeutic genes directly into brain with reasonable safety along with sustained transgene expression. To date, gene therapy for PD that has reached clinical trial evaluation is mainly based on symptomatic approach that involves enzyme replacement strategy and restorative approach that depends on the addition of neurotrophic factors. Mitochondrial dysregulation, such as reduced complex I activity, increased mitochondria-derived reactive oxygen species (ROS) production, ROS-mediated mitochondrial DNA damage, bioenergetic failure, and perturbation of mitochondrial dynamics and mitophagy, has long been implicated in the pathogenesis of PD. Many of mutated genes linked to familial forms of PD affect these mitochondrial features. In this review, we discuss the recent progress that has been made in preclinical development of gene therapy targeting the mitochondrial pathway as disease modifying approach for PD. This review focuses on the potential therapeutic efficacy of candidate genes, including Parkin, PINK1, alpha synuclein, PGC-1 alpha, and anti-apoptotic molecules.
Collapse
Affiliation(s)
- Chi-Jing Choong
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
66
|
Arevalo-Villalobos JI, Rosales-Mendoza S, Zarazua S. Immunotherapies for neurodegenerative diseases: current status and potential of plant-made biopharmaceuticals. Expert Rev Vaccines 2016; 16:151-159. [DOI: 10.1080/14760584.2016.1229602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Jaime I Arevalo-Villalobos
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Sergio Zarazua
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| |
Collapse
|
67
|
Tentillier N, Etzerodt A, Olesen MN, Rizalar FS, Jacobsen J, Bender D, Moestrup SK, Romero-Ramos M. Anti-Inflammatory Modulation of Microglia via CD163-Targeted Glucocorticoids Protects Dopaminergic Neurons in the 6-OHDA Parkinson's Disease Model. J Neurosci 2016; 36:9375-90. [PMID: 27605613 PMCID: PMC6601874 DOI: 10.1523/jneurosci.1636-16.2016] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/22/2016] [Accepted: 07/13/2016] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Increasing evidence supports a decisive role for inflammation in the neurodegenerative process of Parkinson's disease (PD). The immune response in PD seems to involve, not only microglia, but also other immune cells infiltrated into the brain. Indeed, we observed here the infiltration of macrophages, specifically CD163+ macrophages, into the area of neurodegeneration in the 6-hydroxydopamine (6-OHDA) PD model. Therefore, we investigated the therapeutic potential of the infiltrated CD163+ macrophages to modulate local microglia in the brain to achieve neuroprotection. To do so, we designed liposomes targeted for the CD163 receptor to deliver dexamethasone (Dexa) into the CD163+ macrophages in the 6-OHDA PD model. Our data show that a fraction of the CD163-targeted liposomes were carried into the brain after peripheral intravenous injection. The 6-OHDA-lesioned rats that received repeated intravenous CD163-targeted liposomes with Dexa for 3 weeks exhibited better motor performance than the control groups and had minimal glucocorticoid-driven side effects. Furthermore, these animals showed better survival of dopaminergic neurons in substantia nigra and an increased number of microglia expressing major histocompatibility complex II. Therefore, rats receiving CD163-targeted liposomes with Dexa were partially protected against 6-OHDA-induced dopaminergic neurodegeneration, which correlated with a distinctive microglia response. Altogether, our data support the use of macrophages for the modulation of brain neurodegeneration and specifically highlight the potential of CD163-targeted liposomes as a therapeutic tool in PD. SIGNIFICANCE STATEMENT The immune response now evident in the progression of Parkinson's disease comprises both local microglia and other immune cells. We provide evidence that CD163+ macrophages can be a target to modulate brain immune response to achieve neuroprotection in the 6-hydroxydopamine model. To do so, we targeted the CD163+ population, which to a low but significant extent infiltrated in the neurodegenerating area of the brain. Specially designed liposomes targeted for the CD163 receptor were loaded with glucocorticoids and injected peripherally to modify the infiltrated CD163 cells toward an anti-inflammatory profile. This modification of the CD163 population resulted in a distinctive microglial response that correlated with decreased dopaminergic cell death and better motor performance.
Collapse
Affiliation(s)
- Noemie Tentillier
- CNS Disease Modeling Group, NEURODIN, Department of Biomedicine, and
| | | | - Mads N Olesen
- CNS Disease Modeling Group, NEURODIN, Department of Biomedicine, and
| | - F Sila Rizalar
- CNS Disease Modeling Group, NEURODIN, Department of Biomedicine, and
| | - Jan Jacobsen
- Department of Clinical Medicine, PET Center, Aarhus University Hospital, DK-8000 Aarhus C, Denmark, and
| | - Dirk Bender
- Department of Clinical Medicine, PET Center, Aarhus University Hospital, DK-8000 Aarhus C, Denmark, and
| | - Søren K Moestrup
- Department of Biomedicine, and Department of Cancer and Inflammation Research, Syddansk University, DK-5000 Odense, Denmark
| | | |
Collapse
|
68
|
Ingelsson M. Alpha-Synuclein Oligomers-Neurotoxic Molecules in Parkinson's Disease and Other Lewy Body Disorders. Front Neurosci 2016; 10:408. [PMID: 27656123 PMCID: PMC5011129 DOI: 10.3389/fnins.2016.00408] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/23/2016] [Indexed: 12/15/2022] Open
Abstract
Adverse intra- and extracellular effects of toxic α-synuclein are believed to be central to the pathogenesis in Parkinson's disease and other disorders with Lewy body pathology in the nervous system. One of the physiological roles of α-synuclein relates to the regulation of neurotransmitter release at the presynapse, although it is still unclear whether this mechanism depends on the action of monomers or smaller oligomers. As for the pathogenicity, accumulating evidence suggest that prefibrillar species, rather than the deposits per se, are responsible for the toxicity in affected cells. In particular, larger oligomers or protofibrils of α-synuclein have been shown to impair protein degradation as well as the function of several organelles, such as the mitochondria and the endoplasmic reticulum. Accumulating evidence further suggest that oligomers/protofibrils may have a toxic effect on the synapse, which may lead to disrupted electrophysiological properties. In addition, recent data indicate that oligomeric α-synuclein species can spread between cells, either as free-floating proteins or via extracellular vesicles, and thereby act as seeds to propagate disease between interconnected brain regions. Taken together, several lines of evidence suggest that α-synuclein have neurotoxic properties and therefore should be an appropriate molecular target for therapeutic intervention in Parkinson's disease and other disorders with Lewy pathology. In this context, immunotherapy with monoclonal antibodies against α-synuclein oligomers/protofibrils should be a particularly attractive treatment option.
Collapse
Affiliation(s)
- Martin Ingelsson
- Rudbeck Laboratory, Department of Public Health/Geriatrics, Uppsala University Uppsala, Sweden
| |
Collapse
|
69
|
Chen L, Mo M, Li G, Cen L, Wei L, Xiao Y, Chen X, Li S, Yang X, Qu S, Xu P. The biomarkers of immune dysregulation and inflammation response in Parkinson disease. Transl Neurodegener 2016; 5:16. [PMID: 27570618 PMCID: PMC5002148 DOI: 10.1186/s40035-016-0063-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is referring to the multi-systemic α-synucleinopathy with Lewy bodies deposited in midbrain. In ageing, the environmental and genetic factors work together and overactive major histocompatibility complex pathway to regulate immune reactions in central nerve system which resulting in neural degeneration, especially in dopaminergic neurons. As a series of biomarkers, the human leukocyte antigen genes with its related proteomics play cortical roles on the antigen presentation of major histocompatibility complex molecules to stimulate the differentiation of T lymphocytes and i-proteasome activities under their immune response to the PD-related environmental alteration and genetic variation. Furthermore, dopaminergic drugs change the biological characteristic of T lymphatic cells, affect the α-synuclein presentation pathway, and inhibit T lymphatic cells to release cytotoxicity in PD development. Taking together, the serum inflammatory factors and blood T cells are involved in the immune dysregulation of PD and inspected as the potential clinic biomarkers for PD prediction.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, 510120 China
| | - Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, 510120 China
| | - Guangning Li
- Department of Neurology, The Affiliated Huadu Hospital of Southern Medical University, Guangzhou, 510800 China
| | - Luan Cen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, 510080 China
| | - Lei Wei
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, 510120 China
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangdong, 510082 China
| | - Yousheng Xiao
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, 510080 China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, 510120 China
| | - Shaomin Li
- Ann Romney Center for Neurologic Disease, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Xinling Yang
- Department of Neurology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011 China
| | - Shaogang Qu
- Department of Blood Transfusion, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510900 China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, 510120 China
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, 510080 China
| |
Collapse
|
70
|
George S, Brundin P. Immunotherapy in Parkinson's Disease: Micromanaging Alpha-Synuclein Aggregation. JOURNAL OF PARKINSONS DISEASE 2016; 5:413-24. [PMID: 26406122 PMCID: PMC4923719 DOI: 10.3233/jpd-150630] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Currently, several α-synuclein immunotherapies are being tested in experimental Parkinson’s disease models and in clinical trials. Recent research has revealed that α-synuclein is not just an intracellular synaptic protein but also exists extracellularly. Moreover, the transfer of misfolded α-synuclein between cells might be a crucial step in the process leading to a progressive increase in deposition of α-synuclein aggregates throughout the Parkinson’s disease brain. The revelation that α-synuclein is present outside cells has increased the interest in antibody-based therapies and opens up for the notion that microglia might play a key role in retarding Parkinson’s disease progression. The objectives of this review are to describe and contrast the use of active and passive immunotherapy in treating α-synucleinopathies and highlight the likely important role of microglia in clearing misfolded α-synuclein from the extracellular space.
Collapse
Affiliation(s)
| | - Patrik Brundin
- Correspondence to: Patrik Brundin, Van Andel Research Insti-tute, Center for Neurodegenerative Science, 333 Bostwick AvenueNE, Grand Rapids, MI 49503, USA.
Tel.: 616 234 5684; Fax: 616 234 5129.
| |
Collapse
|
71
|
Christiansen JR, Olesen MN, Otzen DE, Romero-Ramos M, Sanchez-Guajardo V. α-Synuclein vaccination modulates regulatory T cell activation and microglia in the absence of brain pathology. J Neuroinflammation 2016; 13:74. [PMID: 27055651 PMCID: PMC4825077 DOI: 10.1186/s12974-016-0532-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/18/2016] [Indexed: 12/22/2022] Open
Abstract
Background Passive and active immunization with α-synuclein has been shown to be neuroprotective in animal models of Parkinson’s disease. We have previously shown that vaccination with α-synuclein, long before α-synuclein-induced brain pathology, prevents striatal degeneration by inducing regulatory T cell infiltration in parenchyma and antibody deposition on α-synuclein overexpressing neurons. However, the effect of peripheral α-synuclein on the immune system is unknown, as are the mechanistic changes induced in the CD4 T cell population during successful neuroprotective animal studies. We have studied the changes induced by vaccination with α-synuclein in the CD4 T cell pool and its impact on brain microglia to understand the immune mechanisms behind successful vaccination strategies in Parkinson’s disease animal models. Methods Mice were immunized with WT or nitrated α-synuclein at a dose equivalent to the one used in our previous successful vaccination strategy and at a higher dose to determine potential dose-dependent effects. Animals were re-vaccinated 4 weeks after and sacrificed 5 days later. These studies were conducted in naive animals in the absence of human α-synuclein expression. Results The CD4 T cell response was modulated by α-synuclein in a dose-dependent manner, in particular the regulatory T cell population. Low-dose α-synuclein induced expansion of naive (Foxp3 + CCR6-CD127lo/neg) and dopamine receptor type D3+ regulatory T cells, as well as an increase in Stat5 protein levels. On the other hand, high dose promoted activation of regulatory T cells (Foxp3CCR6 + CD127lo/neg), which were dopamine receptor D2+D3-, and induced up-regulation of Stat5 and production of anti-α-synuclein antibodies. These effects were specific to the variant of α-synuclein used as the pathology-associated nitrated form induced distinct effects at both doses. The changes observed in the periphery after vaccination with low-dose α-synuclein correlated with an increase in CD154+, CD103+, and CD54+ microglia and the reduction of CD200R+ microglia. This resulted in the induction of a polarized tolerogenic microglia population that was CD200R-CD54CD103CD172a+ (82 % of total microglia). Conclusions We have shown for the first time the mechanisms behind α-synuclein vaccination and, importantly, how we can modulate microglia’s phenotype by regulating the CD4 T cell pool, thus shedding invaluable light on the design of neuroimmunoregulatory therapies for Parkinson’s disease. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0532-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Josefine R Christiansen
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling group, Department of Biomedicine, HEALTH , Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Mads N Olesen
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center - iNANO, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- CNS Disease Modeling group, Department of Biomedicine, HEALTH , Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark. .,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
72
|
Ugen KE, Lin X, Bai G, Liang Z, Cai J, Li K, Song S, Cao C, Sanchez-Ramos J. Evaluation of an α synuclein sensitized dendritic cell based vaccine in a transgenic mouse model of Parkinson disease. Hum Vaccin Immunother 2016; 11:922-30. [PMID: 25714663 DOI: 10.1080/21645515.2015.1012033] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In order to develop a cell-based vaccine against the Parkinson disease (PD) associated protein α-synuclein (α-Syn) 3 peptides were synthesized based upon predicted B cell epitopes within the full length α-Syn protein sequence. These peptide fragments as well as the full length recombinant human α-Syn (rh- α-Syn) protein were used to sensitize mouse bone marrow-derived dendritic cells (DC) ex vivo, followed by intravenous delivery of these sensitized DCs into transgenic (Tg) mice expressing the human A53T variant of α-Syn. ELISA analysis and testing of behavioral locomotor function by rotometry were performed on all mice after the 5th vaccination as well as just prior to euthanasia. The results indicated that vaccination with peptide sensitized DCs (PSDC) as well as DCs sensitized by rh-α-Syn induced specific anti-α-Syn antibodies in all immunized mice. In terms of rotometry performance, a measure of locomotor activity correlated to brain dopamine levels, mice vaccinated with PSDC or rh- α-Syn sensitized DCs performed significantly better than non-vaccinated Tg control mice during the final assessment (i.e. at 17 months of age) before euthanasia. As well, measurement of levels of brain IL-1α, a cytokine hypothesized to be associated with neuroinflammation, demonstrated that this proinflammatory molecule was significantly reduced in the PSDC and rh- α-Syn sensitized DC vaccinated mice compared to the non-vaccinated Tg control group. Overall, α-Syn antigen-sensitized DC vaccination was effective in generating specific anti- α-Syn antibodies and improved locomotor function without eliciting an apparent general inflammatory response, indicating that this strategy may be a safe and effective treatment for PD.
Collapse
Affiliation(s)
- Kenneth E Ugen
- a Department of Molecular Medicine ; University of South Florida; Morsani College of Medicine ; Tampa , FL USA
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Brandel JP, Corbillé AG, Derkinderen P, Haïk S. [Is Parkinson's disease a prion disease?]. Rev Neurol (Paris) 2015; 171:812-24. [PMID: 26563663 PMCID: PMC7111738 DOI: 10.1016/j.neurol.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023]
Abstract
The accumulation of a specific protein in aggregated form is a common phenomenon in human neurodegenerative diseases. In Parkinson's disease, this protein is α-synuclein which is a neuronal protein of 143 amino acids. With a monomeric conformation in solution, it also has a natural capacity to aggregate into amyloid structures (dimers, oligomers, fibrils and Lewy bodies or neurites). It therefore fulfils the characteristics of a prion protein (different conformations, seeding and spreading). In vitro and in vivo experimental evidence in transgenic and wild animals indicates a prion-like propagation of Parkinson's disease. The sequential and predictive distribution of α-synuclein demonstrated by Braak et al. and its correlation with non-motor signs are consistent with the prion-like progression. Although the triggering factor causing the misfolding and aggregation of the target protein is unknown, Parkinson's disease is a highly relevant model for the study of these mechanisms and also to test specific treatments targeting the assemblies of α-synuclein and propagation from pre-motor phase of the disease. Despite this prion-like progression, there is currently no argument indicating a risk of human transmission of Parkinson's disease.
Collapse
Affiliation(s)
- J-P Brandel
- Inserm U 1127, CNRS UMR 7225, Sorbonne universités, UPMC University Paris 06 UMR S 1127, institut du cerveau et de la mœlle épinière, ICM, 75013 Paris, France; Cellule nationale de référence des maladies de Creutzfeldt-Jakob, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France; Centre national de référence des agents transmissibles non conventionnels, 75013 Paris, France; Unité James-Parkinson, Fondation ophtalmologique Rothschild, 75019 Paris, France.
| | - A-G Corbillé
- Département de neurologie, CHU de Nantes, 44093 Nantes, France; Inserm, U913, 44093 Nantes, France
| | - P Derkinderen
- Département de neurologie, CHU de Nantes, 44093 Nantes, France; Inserm, U913, 44093 Nantes, France
| | - S Haïk
- Inserm U 1127, CNRS UMR 7225, Sorbonne universités, UPMC University Paris 06 UMR S 1127, institut du cerveau et de la mœlle épinière, ICM, 75013 Paris, France; Cellule nationale de référence des maladies de Creutzfeldt-Jakob, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France; Centre national de référence des agents transmissibles non conventionnels, 75013 Paris, France
| |
Collapse
|
74
|
Olson KE, Gendelman HE. Immunomodulation as a neuroprotective and therapeutic strategy for Parkinson's disease. Curr Opin Pharmacol 2015; 26:87-95. [PMID: 26571205 DOI: 10.1016/j.coph.2015.10.006] [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] [Received: 09/25/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 01/06/2023]
Abstract
While immune control is associated with nigrostriatal neuroprotection for Parkinson's disease, direct cause and effect relationships have not yet been realized, and modulating the immune system for therapeutic gain has been openly debated. Here, we review how innate and adaptive immunity affect disease pathobiology, and how each could be harnessed for treatment. The overarching idea is to employ immunopharmacologics as neuroprotective strategies for disease. The aim of the current work is to review disease-modifying treatments that are currently being developed as neuroprotective strategies for PD in experimental animal models and for human disease translation. The long-term goal of this research is to effectively harness the immune system to slow or prevent PD pathobiology.
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
75
|
Labrador‐Garrido A, Cejudo‐Guillén M, Daturpalli S, Leal MM, Klippstein R, De Genst EJ, Villadiego J, Toledo‐Aral JJ, Dobson CM, Jackson SE, Pozo D, Roodveldt C. Chaperome screening leads to identification of Grp94/Gp96 and FKBP4/52 as modulators of the α‐synuclein‐elicited immune response. FASEB J 2015; 30:564-77. [DOI: 10.1096/fj.15-275131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/21/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Adahir Labrador‐Garrido
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Marta Cejudo‐Guillén
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Soumya Daturpalli
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - María M. Leal
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
| | - Rebecca Klippstein
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Erwin J. De Genst
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - Javier Villadiego
- Department of Medical Physiology and BiophysicsSchool of MedicineUniversity of SevilleSevilleSpain
- Institute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocioConsejo Superior de Investigaciones Científicas (CSIC)University of SevilleSevilleSpain
- Centers for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)SevilleSpain
| | - Juan J. Toledo‐Aral
- Department of Medical Physiology and BiophysicsSchool of MedicineUniversity of SevilleSevilleSpain
- Institute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocioConsejo Superior de Investigaciones Científicas (CSIC)University of SevilleSevilleSpain
- Centers for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)SevilleSpain
| | | | - Sophie E. Jackson
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - David Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Cintia Roodveldt
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
| |
Collapse
|
76
|
Schneeberger A, Tierney L, Mandler M. Active immunization therapies for Parkinson's disease and multiple system atrophy. Mov Disord 2015; 31:214-24. [PMID: 26260853 DOI: 10.1002/mds.26377] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/16/2015] [Accepted: 07/20/2015] [Indexed: 01/06/2023] Open
Abstract
Vaccination is increasingly being investigated as a potential treatment for synucleinopathies, a group of neurodegenerative diseases including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies associated with α-synuclein pathology. All lack a causal therapy. Development of novel, disease-altering treatment strategies is urgently needed. Vaccination has positioned itself as a prime strategy for addressing these diseases because it is broadly applicable, requires infrequent administration, and maintains low production costs for treating a large population or as a preventive measure. Current evidence points to a causal role of misfolded α-synuclein in the development and progression of synucleinopathies. In the past decade, significant progress in active immunization against α-synuclein has been shown both in preclinical animal models and in early clinical development. In this review, we describe the state-of-the-art in active immunization approaches to synucleinopathies, with a focus on advances in Parkinson's disease (PD) and multiple-system atrophy (MSA). We first review preclinical animal models, highlighting their progress in translation to the clinical setting. We then discuss current clinical applications, stressing different approaches taken to address α-synuclein pathology. Finally, we address challenges, trends, and future perspectives of current vaccination programs.
Collapse
|
77
|
Chen Y, Qi B, Xu W, Ma B, Li L, Chen Q, Qian W, Liu X, Qu H. Clinical correlation of peripheral CD4+‑cell sub‑sets, their imbalance and Parkinson's disease. Mol Med Rep 2015; 12:6105-11. [PMID: 26239429 DOI: 10.3892/mmr.2015.4136] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 06/18/2015] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence suggests that the peripheral immune system has an active role in the progression of Parkinson's disease (PD). The finding of T‑helper (Th; CD4+) cells infiltrating into the substantia nigra in PD patients demonstrated that Th cells are involved in PD. However, the association between peripheral T‑helper cell sub‑sets (Th1, Th2, Treg and Th17) and the sub‑set balance (Th1/Th2 and Th17/Treg) and PD has remained elusive. In the present study, sixty PD patients of the First Affiliated Hospital of Bengbu Medical College as well as 40 age‑ and environment‑matched healthy individuals were enrolled. The fraction of CD4+ T cells in the peripheral blood was assessed by automated hematology analysis and its sub‑sets (Thl, Th2, Thl7, Treg) were quantified by flow cytometry. The results showed that in the PD group, the proportion of Th1 and Th17 cells was increased, while that of Th2 and Treg cells was decreased. Compared with the control group, the Th1/Th2 and Th17/Treg ratios were significantly enhanced, and shifted towards Th1 and Th17, respectively. Furthermore, this Th1‑type response (Th1/Th2 balance shifting towards Th1) were associated with motor function scores determined by Unified Parkinson's Disease Rating Scale III (UPDRS‑III) scores. However, no correlation was found between the change in the Th17/Treg cell balance (Th17/Treg balance shifting towards Th1) and UPDRS‑III scores. These data supported that chronic immune stimulation, specifically CD4+‑cell sub‑set imbalance, is linked to PD pathobiology and disease severity. CD4+‑cell sub‑sets and their imbalance may therefore represent novel biomarkers or therapeutic targets for PD.
Collapse
Affiliation(s)
- Yuhua Chen
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Benquan Qi
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Wenfang Xu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Bo Ma
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Li Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Qiming Chen
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Weidong Qian
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Xiaolin Liu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Hongdang Qu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
78
|
Brundin P, Atkin G, Lamberts JT. Basic science breaks through: New therapeutic advances in Parkinson's disease. Mov Disord 2015; 30:1521-7. [PMID: 26177603 DOI: 10.1002/mds.26332] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/13/2015] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is typically associated with progressive motor dysfunction, although PD patients also exhibit a variety of non-motor symptoms. The neuropathological hallmark of PD is intraneuronal inclusions containing primarily α-Synuclein (α-Syn), and several lines of evidence point to α-Syn as a key contributor to disease progression. Thus, basic research in the field of PD is largely focused on understanding the pathogenic properties of α-Syn. Over the past 2 y, these studies helped to identify several novel therapeutic strategies that have the potential to slow PD progression; such strategies include sequestration of extracellular α-Syn through immunotherapy, reduction of α-Syn multimerization or intracellular toxicity, and attenuation of the neuroinflammatory response. This review describes these and other putative therapeutic strategies, together with the basic science research that led to their identification. The current breadth of novel targets for the treatment of PD warrants cautious optimism in the fight against this devastating disease.
Collapse
Affiliation(s)
- Patrik Brundin
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Graham Atkin
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer T Lamberts
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA.,College of Pharmacy, Ferris State University, Big Rapids, Michigan, USA
| |
Collapse
|
79
|
Regulation of the Neurodegenerative Process Associated to Parkinson's Disease by CD4+ T-cells. J Neuroimmune Pharmacol 2015; 10:561-75. [PMID: 26018603 DOI: 10.1007/s11481-015-9618-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/19/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation constitutes a fundamental process involved in the physiopathology of Parkinson's disease (PD). Microglial cells play a central role in the outcome of neuroinflammation and consequent neurodegeneration of dopaminergic neurons in the substantia nigra. Current evidence indicates that CD4+ T-cells infiltrate the central nervous system (CNS) in PD, where they play a critical role determining the functional phenotype of microglia, thus regulating the progression of the neurodegenerative process. Here, we first analysed the pathogenic role of inflammatory phenotypes and the beneficial role of anti-inflammatory phenotypes of encephalitogenic CD4+ T-cells involved in the physiopathology of PD. Next, we discussed how alterations of neurotransmitter levels observed in the basal ganglia throughout the time course of PD progression could be strongly affecting the behaviour of encephalitogenic CD4+ T-cells and thereby the outcome of the neuroinflammatory process and the consequent neurodegeneration of dopaminergic neurons. Afterward, we integrated the evidence indicating the involvement of an antigen-specific immune response mediated by T-cells and B-cells against CNS-derived self-constituents in PD. Consistent with the involvement of a relevant autoimmune component in PD, we also reviewed the polymorphisms of both, class I and class II major histocompatibility complexes, associated to the risk of PD. Overall, this study gives an overview of how an autoimmune component involved in PD plays a fundamental role in the progression of the neurodegenerative process.
Collapse
|
80
|
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
|
81
|
Labrador-Garrido A, Cejudo-Guillén M, Klippstein R, De Genst EJ, Tomas-Gallardo L, Leal MM, Villadiego J, Toledo-Aral JJ, Dobson CM, Pozo D, Roodveldt C. Chaperoned amyloid proteins for immune manipulation: α-Synuclein/Hsp70 shifts immunity toward a modulatory phenotype. IMMUNITY INFLAMMATION AND DISEASE 2014; 2:226-38. [PMID: 25866630 PMCID: PMC4386917 DOI: 10.1002/iid3.39] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/23/2014] [Accepted: 10/20/2014] [Indexed: 01/02/2023]
Abstract
α-Synuclein (αSyn) is a 140-residue amyloid-forming protein whose aggregation is linked to Parkinson's disease (PD). It has also been found to play a critical role in the immune imbalance that accompanies disease progression, a characteristic that has prompted the search for an effective αSyn-based immunotherapy. In this study, we have simultaneously exploited two important features of certain heat-shock proteins (HSPs): their classical “chaperone” activities and their recently discovered and diverse “immunoactive” properties. In particular, we have explored the immune response elicited by immunization of C57BL/6 mice with an αSyn/Hsp70 protein combination in the absence of added adjuvant. Our results show differential effects for mice immunized with the αSyn/Hsp70 complex, including a restrained αSyn-specific (IgM and IgG) humoral response as well as minimized alterations in the Treg (CD4+CD25+Foxp3+) and Teff (CD4+Foxp3−) cell populations, as opposed to significant changes in mice immunized with αSyn and Hsp70 alone. Furthermore, in vitro-stimulated splenocytes from immunized mice showed the lowest relative response against αSyn challenge for the “αSyn/Hsp70” experimental group as measured by IFN-γ and IL-17 secretion, and higher IL-10 levels when stimulated with LPS. Finally, serum levels of Th1-cytokine IFN-γ and immunomodulatory IL-10 indicated a unique shift toward an immunomodulatory/immunoprotective phenotype in mice immunized with the αSyn/Hsp70 complex. Overall, we propose the use of functional “HSP-chaperoned amyloid/aggregating proteins” generated with appropriate HSP-substrate protein combinations, such as the αSyn/Hsp70 complex, as a novel strategy for immune-based intervention against synucleinopathies and other amyloid or “misfolding” neurodegenerative disorders.
Collapse
Affiliation(s)
- Adahir Labrador-Garrido
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Marta Cejudo-Guillén
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Rebecca Klippstein
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | | | | | - María M Leal
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain
| | - Javier Villadiego
- IBiS Institute of Biomedicine of Seville, University Hospital Virgen del Rocío-CSIC-University of Seville Spain ; Department of Medical Physiology and Biophysics School of Medicine, University of Seville Spain ; CIBERNED, Centers for Networked Biomedical Research in Neurodegenerative Diseases Spain
| | - Juan J Toledo-Aral
- IBiS Institute of Biomedicine of Seville, University Hospital Virgen del Rocío-CSIC-University of Seville Spain ; Department of Medical Physiology and Biophysics School of Medicine, University of Seville Spain ; CIBERNED, Centers for Networked Biomedical Research in Neurodegenerative Diseases Spain
| | | | - David Pozo
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Cintia Roodveldt
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain
| |
Collapse
|
82
|
Lindström V, Ihse E, Fagerqvist T, Bergström J, Nordström E, Möller C, Lannfelt L, Ingelsson M. Immunotherapy targeting α-synuclein, with relevance for future treatment of Parkinson's disease and other Lewy body disorders. Immunotherapy 2014; 6:141-53. [PMID: 24491088 DOI: 10.2217/imt.13.162] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy targeting α-synuclein has evolved as a potential therapeutic strategy for neurodegenerative diseases, such as Parkinson's disease, and initial studies on cellular and animal models have shown promising results. α-synuclein vaccination of transgenic mice reduced the number of brain inclusions, whereas passive immunization studies demonstrated that antibodies against the C-terminus of α-synuclein can pass the blood-brain barrier and affect the pathology. In addition, preliminary evidence suggests that transgenic mice treated with an antibody directed against α-synuclein oligomers/protofibrils resulted in reduced levels of such species in the CNS. The underlying mechanisms of immunotherapy are not yet fully understood, but may include antibody-mediated clearance of pre-existing aggregates, prevention of protein propagation between cells and microglia-dependent protein clearance. Thus, immunotherapy targeting α-synuclein holds promise, but needs to be further developed as a future disease-modifying treatment in Parkinson's disease and other α-synucleinopathies.
Collapse
Affiliation(s)
- Veronica Lindström
- Department of Public Health/Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Jones TB. Lymphocytes and autoimmunity after spinal cord injury. Exp Neurol 2014; 258:78-90. [PMID: 25017889 DOI: 10.1016/j.expneurol.2014.03.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/23/2022]
Abstract
Over the past 15 years an immense amount of data has accumulated regarding the infiltration and activation of lymphocytes in the traumatized spinal cord. Although the impact of the intraspinal accumulation of lymphocytes is still unclear, modulation of the adaptive immune response via active and passive vaccination is being evaluated for its preclinical efficacy in improving the outcome for spinal-injured individuals. The complexity of the interaction between the nervous and the immune systems is highlighted in the contradictions that appear in response to these modulations. Current evidence regarding augmentation and inhibition of the adaptive immune response to spinal cord injury is reviewed with an aim toward reconciling conflicting data and providing consensus issues that may be exploited in future therapies. Opportunities such an approach may provide are highlighted as well as the obstacles that must be overcome before such approaches can be translated into clinical trials.
Collapse
Affiliation(s)
- T Bucky Jones
- Department of Anatomy, Arizona College of Medicine, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
84
|
Romero-Ramos M, von Euler Chelpin M, Sanchez-Guajardo V. Vaccination strategies for Parkinson disease: induction of a swift attack or raising tolerance? Hum Vaccin Immunother 2014; 10:852-67. [PMID: 24670306 DOI: 10.4161/hv.28578] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson disease is the second most common neurodegenerative disease in the world, but there is currently no available cure for it. Current treatments only alleviate some of the symptoms for a few years, but they become ineffective in the long run and do not stop the disease. Therefore it is of outmost importance to develop therapeutic strategies that can prevent, stop, or cure Parkinson disease. A very promising target for these therapies is the peripheral immune system due to its probable involvement in the disease and its potential as a tool to modulate neuroinflammation. But for such strategies to be successful, we need to understand the particular state of the peripheral immune system during Parkinson disease in order to avoid its weaknesses. In this review we examine the available data regarding how dopamine regulates the peripheral immune system and how this regulation is affected in Parkinson disease; the specific cytokine profiles observed during disease progression and the alterations documented to date in patients' peripheral blood mononuclear cells. We also review the different strategies used in Parkinson disease animal models to modulate the adaptive immune response to salvage dopaminergic neurons from cell death. After analyzing the evidence, we hypothesize the need to prime the immune system to restore natural tolerance against α-synuclein in Parkinson disease, including at the same time B and T cells, so that T cells can reprogram microglia activation to a beneficial pattern and B cell/IgG can help neurons cope with the pathological forms of α-synuclein.
Collapse
Affiliation(s)
- Marina Romero-Ramos
- CNS disease modeling group; Department of Biomedicine; Aarhus University; Aarhus, Denmark; NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| | - Marianne von Euler Chelpin
- CNS disease modeling group; Department of Biomedicine; Aarhus University; Aarhus, Denmark; NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark; Neuroimmunology of Degenerative Diseases group; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark; Neuroimmunology of Degenerative Diseases group; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| |
Collapse
|
85
|
Sanchez-Guajardo V, Barnum C, Tansey M, Romero-Ramos M. Neuroimmunological processes in Parkinson's disease and their relation to α-synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro 2013; 5:113-39. [PMID: 23506036 PMCID: PMC3639751 DOI: 10.1042/an20120066] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 03/15/2013] [Accepted: 03/19/2013] [Indexed: 12/15/2022] Open
Abstract
The role of neuroinflammation and the adaptive immune system in PD (Parkinson's disease) has been the subject of intense investigation in recent years, both in animal models of parkinsonism and in post-mortem PD brains. However, how these processes relate to and modulate α-syn (α-synuclein) pathology and microglia activation is still poorly understood. Specifically, how the peripheral immune system interacts, regulates and/or is induced by neuroinflammatory processes taking place during PD is still undetermined. We present herein a comprehensive review of the features and impact that neuroinflamation has on neurodegeneration in different animal models of nigral cell death, how this neuroinflammation relates to microglia activation and the way microglia respond to α-syn in vivo. We also discuss a possible role for the peripheral immune system in animal models of parkinsonism, how these findings relate to the state of microglia activation observed in these animal models and how these findings compare with what has been observed in humans with PD. Together, the available data points to the need for development of dual therapeutic strategies that modulate microglia activation to change not only the way microglia interact with the peripheral immune system, but also to modulate the manner in which microglia respond to encounters with α-syn. Lastly, we discuss the immune-modulatory strategies currently under investigation in animal models of parkinsonism and the degree to which one might expect their outcomes to translate faithfully to a clinical setting.
Collapse
Key Words
- lymphocytes
- m1/m2 phenotype
- microglia
- neuroinflammation
- parkinson’s disease
- α-synuclein
- 6-ohda, 6-hydroxydopamine
- ad, alzheimer’s disease
- apc, antigen-presenting cell
- α-syn, α-synuclein
- bbb, brain–blood barrier
- bcg, bacille calmette–guérin
- bm, bone marrow
- cfa, complete freund’s adjuvant
- cm, conditioned media
- cns, central nervous system
- cox, cyclooxygenase
- cr, complement receptor
- csf, cerebrospinal fluid
- da, dopamine
- eae, experimental autoimmune encephalomyelitis
- ga, galatiramer acetate
- gdnf, glial-derived neurotrophic factor
- gfp, green fluorescent protein
- hla-dr, human leucocyte antigen type dr
- ifnγ, interferon γ
- igg, immunoglobulin g
- il, interleukin
- inos, inducible nitric oxide synthase
- lamp, lysosome-associated membrane protein
- lb, lewy body
- lps, lipopolysaccharide
- mhc, major histocompatibility complex
- mptp, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- nfκb, nuclear factor κb
- nk, natural killer
- no, nitric oxide
- pd, parkinson’s disease
- pet, positron-emission tomography
- prp, prion protein
- raav, recombinant adeno-associated virus
- rns, reactive nitrogen species
- ros, reactive oxygen species
- sn, substantia nigra
- snp, single nucleotide polymorphism
- tcr, t-cell receptor
- tgfβ, tumour growth factor β
- th, tyrosine hydroxylase
- th1, t helper 1
- tlr, toll-like receptor
- tnf, tumour necrosis factor
- treg, regulatory t-cell
- vip, vasoactive intestinal peptide
- wt, wild-type
Collapse
Affiliation(s)
- Vanesa Sanchez-Guajardo
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christopher J. Barnum
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Malú G. Tansey
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Marina Romero-Ramos
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|