1
|
Saadh MJ, Ahmed HH, Kareem RA, Sanghvi G, Ganesan S, Agarwal M, Kaur P, Taher WM, Alwan M, Jawad MJ, Hamad AK. Short-chain fatty acids in Huntington's disease: Mechanisms of action and their therapeutic implications. Pharmacol Biochem Behav 2025; 249:173972. [PMID: 39983928 DOI: 10.1016/j.pbb.2025.173972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor dysfunction, cognitive decline, and emotional instability, primarily resulting from the abnormal accumulation of mutant huntingtin protein. Growing research highlights the role of intestinal microbiota and their metabolites, particularly short-chain fatty acids (SCFAs), in modulating HD progression. SCFAs, including acetate, propionate, and butyrate, are produced by gut bacteria through dietary fiber fermentation and are recognized for their neuroprotective properties. Evidence suggests that SCFAs regulate neuroinflammation, neuronal communication, and metabolic functions within the central nervous system (CNS). In HD, these compounds may support neuronal health, reduce oxidative stress, and enhance blood-brain barrier (BBB) integrity. Their mechanisms of action involve binding to G-protein-coupled receptors (GPCRs) and modulating gene expression through epigenetic pathways, underscoring their therapeutic potential. This analysis examines the significance of SCFAs in HD, emphasizing the gut-brain axis and the benefits of dietary interventions aimed at modifying gut microbiota composition and promoting SCFA production. Further research into these pathways may pave the way for novel HD management strategies and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mohit Agarwal
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University, Rajasthan, Jaipur,302131, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
2
|
Fung JN, Lee JD, Adam R, O'Sullivan JD, Woodruff TM. Peripheral and central elevation of IL-8 in patients with Huntington's disease. Mol Immunol 2025; 179:84-93. [PMID: 39923663 DOI: 10.1016/j.molimm.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Huntington's Disease (HD) is a debilitating neurodegenerative condition characterized by motor, cognitive and psychiatric abnormalities. Immune hyperactivity and dysregulation are common in HD. In addition to the central nervous system, HD patients exhibit systemic innate immune activation and inflammation, which has been shown to contribute to the pathogenic effects of the Huntingtin gene mutation. Upregulation of inflammatory mediators including interferon gamma (IFN-γ) and interleukin (IL)-8 has been observed in animal Huntington's disease models. However, studies on HD patients remain limited. METHODS In this study, serum samples from 58 HD patients and 59 age- and gender-matched healthy control individuals were analysed using a bead-based assay, that enabled simultaneous measurement of 13 cytokines and chemokines. Additionally, publicly available transcriptomic data from brain tissues of HD patients and controls were examined. RESULTS Our results confirm that IL-8 protein levels are significantly higher in HD patients compared to non-HD controls, with the highest levels observed in the moderate HD group. In the control group, we found significant positive correlations between IL-8 levels and both IL-17A and IL-10. However, these correlations were not observed in HD patients, where IL-8 levels were notably positively correlated with pro-inflammatory markers including IFNγ and IL-23. Interestingly, IL-17A levels demonstrated a negative correlation with disease parameters, including CAG trinucleotide repeat expansion and disease burden score. Furthermore, cytokines and chemokines such as IFNγ and monocyte chemoattractant protein 1 (MCP-1; CCL2) demonstrated positive correlations with the same disease parameters. In-depth analysis of publicly available bulk RNAseq, and single-nucleus RNA-sequencing (snRNAseq) data from two key HD-affected brain regions- the prefrontal cortex and striatum revealed that IL-8 expression is significantly increased in cortex samples from individuals with HD compared to non-HD controls. Moreover, snRNAseq data in the striatum showed higher IL-8 expression in HD patients than in non-HD controls, with a predominant expression in microglia. CONCLUSION Overall, our findings support an upregulation of IL-8 in patients with HD, evident in both central degenerating brain regions, and peripheral blood samples. We identified unique immunological signatures associated with the severity of HD and provide potential biomarkers that may reflect immune-pathological mechanisms in HD patients.
Collapse
Affiliation(s)
- Jenny N Fung
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Robert Adam
- Neurology Department, Royal Brisbane and Women's Hospital, Herston, Brisbane, QLD 4029, Australia; Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Brisbane, QLD 4029, Australia
| | - John D O'Sullivan
- Neurology Department, Royal Brisbane and Women's Hospital, Herston, Brisbane, QLD 4029, Australia; Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Brisbane, QLD 4029, Australia.
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia; Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
3
|
Ekwudo MN, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington's disease: pathogenic mechanisms and therapeutic targets. FEBS J 2025; 292:1282-1315. [PMID: 38426291 PMCID: PMC11927060 DOI: 10.1111/febs.17102] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Huntington's disease (HD) is a currently incurable neurogenerative disorder and is typically characterized by progressive movement disorder (including chorea), cognitive deficits (culminating in dementia), psychiatric abnormalities (the most common of which is depression), and peripheral symptoms (including gastrointestinal dysfunction). There are currently no approved disease-modifying therapies available for HD, with death usually occurring approximately 10-25 years after onset, but some therapies hold promising potential. HD subjects are often burdened by chronic diarrhea, constipation, esophageal and gastric inflammation, and a susceptibility to diabetes. Our understanding of the microbiota-gut-brain axis in HD is in its infancy and growing evidence from preclinical and clinical studies suggests a role of gut microbial population imbalance (gut dysbiosis) in HD pathophysiology. The gut and the brain can communicate through the enteric nervous system, immune system, vagus nerve, and microbiota-derived-metabolites including short-chain fatty acids, bile acids, and branched-chain amino acids. This review summarizes supporting evidence demonstrating the alterations in bacterial and fungal composition that may be associated with HD. We focus on mechanisms through which gut dysbiosis may compromise brain and gut health, thus triggering neuroinflammatory responses, and further highlight outcomes of attempts to modulate the gut microbiota as promising therapeutic strategies for HD. Ultimately, we discuss the dearth of data and the need for more longitudinal and translational studies in this nascent field. We suggest future directions to improve our understanding of the association between gut microbes and the pathogenesis of HD, and other 'brain and body disorders'.
Collapse
Affiliation(s)
- Millicent N. Ekwudo
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
- Department of Anatomy and PhysiologyUniversity of MelbourneParkvilleAustralia
| |
Collapse
|
4
|
Cicardi ME, Trotti D. C9orf72 role in myeloid cells: new perspectives in the investigation of the neuro-immune crosstalk in amyotrophic lateral sclerosis and frontotemporal dementia. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:120. [PMID: 39817235 PMCID: PMC11729807 DOI: 10.21037/atm-24-86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/19/2024] [Indexed: 01/18/2025]
Affiliation(s)
- Maria Elena Cicardi
- Department of Neuroscience, Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Davide Trotti
- Department of Neuroscience, Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
5
|
Li X, Tong H, Xu S, Zhou G, Yang T, Yin S, Yang S, Li X, Li S. Neuroinflammatory Proteins in Huntington's Disease: Insights into Mechanisms, Diagnosis, and Therapeutic Implications. Int J Mol Sci 2024; 25:11787. [PMID: 39519337 PMCID: PMC11546928 DOI: 10.3390/ijms252111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a CAG tract expansion in the huntingtin gene (HTT). HD is characterized by involuntary movements, cognitive decline, and behavioral changes. Pathologically, patients with HD show selective striatal neuronal vulnerability at the early disease stage, although the mutant protein is ubiquitously expressed. Activation of the immune system and glial cell-mediated neuroinflammatory responses are early pathological features and have been found in all neurodegenerative diseases (NDDs), including HD. However, the role of inflammation in HD, as well as its therapeutic significance, has been less extensively studied compared to other NDDs. This review highlights the significantly elevated levels of inflammatory proteins and cellular markers observed in various HD animal models and HD patient tissues, emphasizing the critical roles of microglia, astrocytes, and oligodendrocytes in mediating neuroinflammation in HD. Moreover, it expands on recent discoveries related to the peripheral immune system's involvement in HD. Although current immunomodulatory treatments and inflammatory biomarkers for adjunctive diagnosis in HD are limited, targeting inflammation in combination with other therapies, along with comprehensive personalized treatment approaches, shows promising therapeutic potential.
Collapse
Affiliation(s)
- Xinhui Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Huichun Tong
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Gongke Zhou
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Tianqi Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shurui Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Sitong Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Xiaojiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| |
Collapse
|
6
|
Field SE, Curle AJ, Barker RA. Inflammation and Huntington's disease - a neglected therapeutic target? Expert Opin Investig Drugs 2024; 33:451-467. [PMID: 38758356 DOI: 10.1080/13543784.2024.2348738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Huntington's Disease (HD) is a genetic neurodegenerative disease for which there is currently no disease-modifying treatment. One of several underlying mechanisms proposed to be involved in HD pathogenesis is inflammation; there is now accumulating evidence that the immune system may play an integral role in disease pathology and progression. As such, modulation of the immune system could be a potential therapeutic target for HD. AREAS COVERED To date, the number of trials targeting immune aspects of HD has been limited. However, targeting it, may have great advantages over other therapeutic areas, given that many drugs already exist that have actions in this system coupled to the fact that inflammation can be measured both peripherally and, to some extent, centrally using CSF and PET imaging. In this review, we look at evidence that the immune system and the newly emerging area of the microbiome are altered in HD patients, and then present and discuss clinical trials that have targeted different parts of the immune system. EXPERT OPINION We then conclude by discussing how this field might develop going forward, focusing on the role of imaging and other biomarkers to monitor central immune activation and response to novel treatments in HD.
Collapse
Affiliation(s)
- Sophie E Field
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Annabel J Curle
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Burtscher J, Strasser B, Pepe G, Burtscher M, Kopp M, Di Pardo A, Maglione V, Khamoui AV. Brain-Periphery Interactions in Huntington's Disease: Mediators and Lifestyle Interventions. Int J Mol Sci 2024; 25:4696. [PMID: 38731912 PMCID: PMC11083237 DOI: 10.3390/ijms25094696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Prominent pathological features of Huntington's disease (HD) are aggregations of mutated Huntingtin protein (mHtt) in the brain and neurodegeneration, which causes characteristic motor (such as chorea and dystonia) and non-motor symptoms. However, the numerous systemic and peripheral deficits in HD have gained increasing attention recently, since those factors likely modulate disease progression, including brain pathology. While whole-body metabolic abnormalities and organ-specific pathologies in HD have been relatively well described, the potential mediators of compromised inter-organ communication in HD have been insufficiently characterized. Therefore, we applied an exploratory literature search to identify such mediators. Unsurprisingly, dysregulation of inflammatory factors, circulating mHtt, and many other messenger molecules (hormones, lipids, RNAs) were found that suggest impaired inter-organ communication, including of the gut-brain and muscle-brain axis. Based on these findings, we aimed to assess the risks and potentials of lifestyle interventions that are thought to improve communication across these axes: dietary strategies and exercise. We conclude that appropriate lifestyle interventions have great potential to reduce symptoms and potentially modify disease progression (possibly via improving inter-organ signaling) in HD. However, impaired systemic metabolism and peripheral symptoms warrant particular care in the design of dietary and exercise programs for people with HD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Barbara Strasser
- Ludwig Boltzmann Institute for Rehabilitation Research, 1100 Vienna, Austria;
- Faculty of Medicine, Sigmund Freud Private University, 1020 Vienna, Austria
| | - Giuseppe Pepe
- IRCCS Neuromed, 86077 Pozzilli, Italy; (G.P.); (A.D.P.); (V.M.)
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.K.)
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.K.)
| | - Alba Di Pardo
- IRCCS Neuromed, 86077 Pozzilli, Italy; (G.P.); (A.D.P.); (V.M.)
| | | | - Andy V. Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33458, USA;
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
8
|
Niso-Santano M, Fuentes JM, Galluzzi L. Immunological aspects of central neurodegeneration. Cell Discov 2024; 10:41. [PMID: 38594240 PMCID: PMC11004155 DOI: 10.1038/s41421-024-00666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/02/2024] [Indexed: 04/11/2024] Open
Abstract
The etiology of various neurodegenerative disorders that mainly affect the central nervous system including (but not limited to) Alzheimer's disease, Parkinson's disease and Huntington's disease has classically been attributed to neuronal defects that culminate with the loss of specific neuronal populations. However, accumulating evidence suggests that numerous immune effector cells and the products thereof (including cytokines and other soluble mediators) have a major impact on the pathogenesis and/or severity of these and other neurodegenerative syndromes. These observations not only add to our understanding of neurodegenerative conditions but also imply that (at least in some cases) therapeutic strategies targeting immune cells or their products may mediate clinically relevant neuroprotective effects. Here, we critically discuss immunological mechanisms of central neurodegeneration and propose potential strategies to correct neurodegeneration-associated immunological dysfunction with therapeutic purposes.
Collapse
Affiliation(s)
- Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - José M Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Khoshnan A. Gut Microbiota as a Modifier of Huntington's Disease Pathogenesis. J Huntingtons Dis 2024; 13:133-147. [PMID: 38728199 PMCID: PMC11307070 DOI: 10.3233/jhd-240012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2024] [Indexed: 05/12/2024]
Abstract
Huntingtin (HTT) protein is expressed in most cell lineages, and the toxicity of mutant HTT in multiple organs may contribute to the neurological and psychiatric symptoms observed in Huntington's disease (HD). The proteostasis and neurotoxicity of mutant HTT are influenced by the intracellular milieu and responses to environmental signals. Recent research has highlighted a prominent role of gut microbiota in brain and immune system development, aging, and the progression of neurological disorders. Several studies suggest that mutant HTT might disrupt the homeostasis of gut microbiota (known as dysbiosis) and impact the pathogenesis of HD. Dysbiosis has been observed in HD patients, and in animal models of the disease it coincides with mutant HTT aggregation, abnormal behaviors, and reduced lifespan. This review article aims to highlight the potential toxicity of mutant HTT in organs and pathways within the microbiota-gut-immune-central nervous system (CNS) axis. Understanding the functions of Wild-Type (WT) HTT and the toxicity of mutant HTT in these organs and the associated networks may elucidate novel pathogenic pathways, identify biomarkers and peripheral therapeutic targets for HD.
Collapse
Affiliation(s)
- Ali Khoshnan
- Keck School of Medicine, Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Li C, Lin Y, Chen Y, Song X, Zheng X, Li J, He J, Chen X, Huang C, Wang W, Wu J, Wu J, Gao J, Tu Z, Li XJ, Yan S, Li S. A Specific Mini-Intrabody Mediates Lysosome Degradation of Mutant Huntingtin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301120. [PMID: 37688357 PMCID: PMC10625127 DOI: 10.1002/advs.202301120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Accumulation of misfolded proteins leads to many neurodegenerative diseases that can be treated by lowering or removing mutant proteins. Huntington's disease (HD) is characterized by the intracellular accumulation of mutant huntingtin (mHTT) that can be soluble and aggregated in the central nervous system and causes neuronal damage and death. Here, an intracellular antibody (intrabody) fragment is generated that can specifically bind mHTT and link to the lysosome for degradation. It is found that delivery of this peptide by either brain injection or intravenous administration can efficiently clear the soluble and aggregated mHTT by activating the lysosomal degradation pathway, resulting in amelioration of gliosis and dyskinesia in HD knock-in (KI-140Q) mice. These findings suggest that the small intrabody peptide linked to lysosomes can effectively lower mutant proteins and provide a new approach for treating neurodegenerative diseases that are caused by the accumulation of mutant proteins.
Collapse
Affiliation(s)
- Caijuan Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yingqi Lin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yizhi Chen
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao Zheng
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiawei Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jun He
- Institute of Laboratory Animal Science, Jinan University, Guangzhou, 510632, China
| | - Xiusheng Chen
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Chunhui Huang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Wei Wang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jianhao Wu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiaxi Wu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiale Gao
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Zhuchi Tu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
11
|
Wilton DK, Mastro K, Heller MD, Gergits FW, Willing CR, Fahey JB, Frouin A, Daggett A, Gu X, Kim YA, Faull RLM, Jayadev S, Yednock T, Yang XW, Stevens B. Microglia and complement mediate early corticostriatal synapse loss and cognitive dysfunction in Huntington's disease. Nat Med 2023; 29:2866-2884. [PMID: 37814059 PMCID: PMC10667107 DOI: 10.1038/s41591-023-02566-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Huntington's disease (HD) is a devastating monogenic neurodegenerative disease characterized by early, selective pathology in the basal ganglia despite the ubiquitous expression of mutant huntingtin. The molecular mechanisms underlying this region-specific neuronal degeneration and how these relate to the development of early cognitive phenotypes are poorly understood. Here we show that there is selective loss of synaptic connections between the cortex and striatum in postmortem tissue from patients with HD that is associated with the increased activation and localization of complement proteins, innate immune molecules, to these synaptic elements. We also found that levels of these secreted innate immune molecules are elevated in the cerebrospinal fluid of premanifest HD patients and correlate with established measures of disease burden.In preclinical genetic models of HD, we show that complement proteins mediate the selective elimination of corticostriatal synapses at an early stage in disease pathogenesis, marking them for removal by microglia, the brain's resident macrophage population. This process requires mutant huntingtin to be expressed in both cortical and striatal neurons. Inhibition of this complement-dependent elimination mechanism through administration of a therapeutically relevant C1q function-blocking antibody or genetic ablation of a complement receptor on microglia prevented synapse loss, increased excitatory input to the striatum and rescued the early development of visual discrimination learning and cognitive flexibility deficits in these models. Together, our findings implicate microglia and the complement cascade in the selective, early degeneration of corticostriatal synapses and the development of cognitive deficits in presymptomatic HD; they also provide new preclinical data to support complement as a therapeutic target for early intervention.
Collapse
Affiliation(s)
- Daniel K Wilton
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
| | - Kevin Mastro
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Molly D Heller
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Frederick W Gergits
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Carly Rose Willing
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Jaclyn B Fahey
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Arnaud Frouin
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Anthony Daggett
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Xiaofeng Gu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Yejin A Kim
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Richard L M Faull
- Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ted Yednock
- Annexon Biosciences, South San Francisco, CA, USA
| | - X William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Beth Stevens
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
- Stanley Center, Broad Institute, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Gasser J, Gillet G, Valadas JS, Rouvière L, Kotian A, Fan W, Keaney J, Kadiu I. Innate immune activation and aberrant function in the R6/2 mouse model and Huntington's disease iPSC-derived microglia. Front Mol Neurosci 2023; 16:1191324. [PMID: 37415834 PMCID: PMC10319581 DOI: 10.3389/fnmol.2023.1191324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 07/08/2023] Open
Abstract
Huntington's disease (HD) is an inherited autosomal dominant neurodegenerative disease caused by CAG repeats in exon 1 of the HTT gene. A hallmark of HD along with other psychiatric and neurodegenerative diseases is alteration in the neuronal circuitry and synaptic loss. Microglia and peripheral innate immune activation have been reported in pre-symptomatic HD patients; however, what "activation" signifies for microglial and immune function in HD and how it impacts synaptic health remains unclear. In this study we sought to fill these gaps by capturing immune phenotypes and functional activation states of microglia and peripheral immunity in the R6/2 model of HD at pre-symptomatic, symptomatic and end stages of disease. These included characterizations of microglial phenotypes at single cell resolution, morphology, aberrant functions such as surveillance and phagocytosis and their impact on synaptic loss in vitro and ex vivo in R6/2 mouse brain tissue slices. To further understand how relevant the observed aberrant microglial behaviors are to human disease, transcriptomic analysis was performed using HD patient nuclear sequencing data and functional assessments were conducted using induced pluripotent stem cell (iPSC)-derived microglia. Our results show temporal changes in brain infiltration of peripheral lymphoid and myeloid cells, increases in microglial activation markers and phagocytic functions at the pre-symptomatic stages of disease. Increases in microglial surveillance and synaptic uptake parallel significant reduction of spine density in R6/2 mice. These findings were mirrored by an upregulation of gene signatures in the endocytic and migratory pathways in disease-associated microglial subsets in human HD brains, as well as increased phagocytic and migratory functions of iPSC-derived HD microglia. These results collectively suggest that targeting key and specific microglial functions related to synaptic surveillance and pruning may be therapeutically beneficial in attenuating cognitive decline and psychiatric aspects of HD.
Collapse
Affiliation(s)
- Julien Gasser
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Gaelle Gillet
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Jorge S. Valadas
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Laura Rouvière
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Apoorva Kotian
- Development Science, UCB Biopharma SRL, Slough, United Kingdom
| | - Wenqiang Fan
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - James Keaney
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Irena Kadiu
- Neuroinflammation Focus Area, Neuroscience Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| |
Collapse
|
13
|
Pathak D, Sriram K. Molecular Mechanisms Underlying Neuroinflammation Elicited by Occupational Injuries and Toxicants. Int J Mol Sci 2023; 24:2272. [PMID: 36768596 PMCID: PMC9917383 DOI: 10.3390/ijms24032272] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Occupational injuries and toxicant exposures lead to the development of neuroinflammation by activating distinct mechanistic signaling cascades that ultimately culminate in the disruption of neuronal function leading to neurological and neurodegenerative disorders. The entry of toxicants into the brain causes the subsequent activation of glial cells, a response known as 'reactive gliosis'. Reactive glial cells secrete a wide variety of signaling molecules in response to neuronal perturbations and thus play a crucial role in the progression and regulation of central nervous system (CNS) injury. In parallel, the roles of protein phosphorylation and cell signaling in eliciting neuroinflammation are evolving. However, there is limited understanding of the molecular underpinnings associated with toxicant- or occupational injury-mediated neuroinflammation, gliosis, and neurological outcomes. The activation of signaling molecules has biological significance, including the promotion or inhibition of disease mechanisms. Nevertheless, the regulatory mechanisms of synergism or antagonism among intracellular signaling pathways remain elusive. This review highlights the research focusing on the direct interaction between the immune system and the toxicant- or occupational injury-induced gliosis. Specifically, the role of occupational injuries, e.g., trips, slips, and falls resulting in traumatic brain injury, and occupational toxicants, e.g., volatile organic compounds, metals, and nanoparticles/nanomaterials in the development of neuroinflammation and neurological or neurodegenerative diseases are highlighted. Further, this review recapitulates the recent advancement related to the characterization of the molecular mechanisms comprising protein phosphorylation and cell signaling, culminating in neuroinflammation.
Collapse
Affiliation(s)
| | - Krishnan Sriram
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| |
Collapse
|
14
|
Jia Q, Li S, Li XJ, Yin P. Neuroinflammation in Huntington's disease: From animal models to clinical therapeutics. Front Immunol 2022; 13:1088124. [PMID: 36618375 PMCID: PMC9815700 DOI: 10.3389/fimmu.2022.1088124] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease characterized by preferential loss of neurons in the striatum in patients, which leads to motor and cognitive impairments and death that often occurs 10-15 years after the onset of symptoms. The expansion of a glutamine repeat (>36 glutamines) in the N-terminal region of huntingtin (HTT) has been defined as the cause of HD, but the mechanism underlying neuronal death remains unclear. Multiple mechanisms, including inflammation, may jointly contribute to HD pathogenesis. Altered inflammation response is evident even before the onset of classical symptoms of HD. In this review, we summarize the current evidence on immune and inflammatory changes, from HD animal models to clinical phenomenon of patients with HD. The understanding of the impact of inflammation on HD would help develop novel strategies to treat HD.
Collapse
Affiliation(s)
| | | | | | - Peng Yin
- *Correspondence: Xiao-Jiang Li, ; Peng Yin,
| |
Collapse
|
15
|
Puri V, Kanojia N, Sharma A, Huanbutta K, Dheer D, Sangnim T. Natural product-based pharmacological studies for neurological disorders. Front Pharmacol 2022; 13:1011740. [PMID: 36419628 PMCID: PMC9676372 DOI: 10.3389/fphar.2022.1011740] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2023] Open
Abstract
Central nervous system (CNS) disorders and diseases are expected to rise sharply in the coming years, partly because of the world's aging population. Medicines for the treatment of the CNS have not been successfully made. Inadequate knowledge about the brain, pharmacokinetic and dynamic errors in preclinical studies, challenges with clinical trial design, complexity and variety of human brain illnesses, and variations in species are some potential scenarios. Neurodegenerative diseases (NDDs) are multifaceted and lack identifiable etiological components, and the drugs developed to treat them did not meet the requirements of those who anticipated treatments. Therefore, there is a great demand for safe and effective natural therapeutic adjuvants. For the treatment of NDDs and other memory-related problems, many herbal and natural items have been used in the Ayurvedic medical system. Anxiety, depression, Parkinson's, and Alzheimer's diseases (AD), as well as a plethora of other neuropsychiatric disorders, may benefit from the use of plant and food-derived chemicals that have antidepressant or antiepileptic properties. We have summarized the present level of knowledge about natural products based on topological evidence, bioinformatics analysis, and translational research in this review. We have also highlighted some clinical research or investigation that will help us select natural products for the treatment of neurological conditions. In the present review, we have explored the potential efficacy of phytoconstituents against neurological diseases. Various evidence-based studies and extensive recent investigations have been included, which will help pharmacologists reduce the progression of neuronal disease.
Collapse
Affiliation(s)
- Vivek Puri
- Chitkara School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Neha Kanojia
- Chitkara School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Ameya Sharma
- Chitkara School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Kampanart Huanbutta
- School of Pharmacy, Eastern Asia University, Rangsit, Pathum Thani, Thailand
| | - Divya Dheer
- Chitkara School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Tanikan Sangnim
- Faculty of Pharmaceutical Sciences, Burapha University, Muang, Chon Buri, Thailand
| |
Collapse
|
16
|
Scarabino D, Veneziano L, Fiore A, Nethisinghe S, Mantuano E, Garcia-Moreno H, Bellucci G, Solanky N, Morello M, Zanni G, Corbo RM, Giunti P. Leukocyte Telomere Length Variability as a Potential Biomarker in Patients with PolyQ Diseases. Antioxidants (Basel) 2022; 11:1436. [PMID: 35892638 PMCID: PMC9332235 DOI: 10.3390/antiox11081436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/02/2022] Open
Abstract
SCA1, SCA2, and SCA3 are the most common forms of SCAs among the polyglutamine disorders, which include Huntington's Disease (HD). We investigated the relationship between leukocyte telomere length (LTL) and the phenotype of SCA1, SCA2, and SCA3, comparing them with HD. The results showed that LTL was significantly reduced in SCA1 and SCA3 patients, while LTL was significantly longer in SCA2 patients. A significant negative relationship between LTL and age was observed in SCA1 but not in SCA2 subjects. LTL of SCA3 patients depend on both patient's age and disease duration. The number of CAG repeats did not affect LTL in the three SCAs. Since LTL is considered an indirect marker of an inflammatory response and oxidative damage, our data suggest that in SCA1 inflammation is present already at an early stage of disease similar to in HD, while in SCA3 inflammation and impaired antioxidative processes are associated with disease progression. Interestingly, in SCA2, contrary to SCA1 and SCA3, the length of leukocyte telomeres does not reduce with age. We have observed that SCAs and HD show a differing behavior in LTL for each subtype, which could constitute relevant biomarkers if confirmed in larger cohorts and longitudinal studies.
Collapse
Affiliation(s)
- Daniela Scarabino
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy
| | - Liana Veneziano
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy;
| | - Alessia Fiore
- Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy; (A.F.); (R.M.C.)
| | - Suran Nethisinghe
- Ataxia Center, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, University College, London WC1N 3BG, UK; (S.N.); (H.G.-M.); (N.S.); (P.G.)
| | - Elide Mantuano
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy;
| | - Hector Garcia-Moreno
- Ataxia Center, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, University College, London WC1N 3BG, UK; (S.N.); (H.G.-M.); (N.S.); (P.G.)
| | - Gianmarco Bellucci
- Department of Neurosciences, Mental Health and Sensory Organs, Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome, 00185 Rome, Italy;
| | - Nita Solanky
- Ataxia Center, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, University College, London WC1N 3BG, UK; (S.N.); (H.G.-M.); (N.S.); (P.G.)
| | - Maria Morello
- Department of Experimental Medicine and Surgery, Tor Vergata University, 00133 Rome, Italy;
| | - Ginevra Zanni
- Unit of Neuromuscolar and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children’s Research Hospital, IRCCS, 00100 Rome, Italy;
| | - Rosa Maria Corbo
- Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy; (A.F.); (R.M.C.)
| | - Paola Giunti
- Ataxia Center, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, University College, London WC1N 3BG, UK; (S.N.); (H.G.-M.); (N.S.); (P.G.)
| |
Collapse
|
17
|
Gómez-Jaramillo L, Cano-Cano F, González-Montelongo MDC, Campos-Caro A, Aguilar-Diosdado M, Arroba AI. A New Perspective on Huntington's Disease: How a Neurological Disorder Influences the Peripheral Tissues. Int J Mol Sci 2022; 23:6089. [PMID: 35682773 PMCID: PMC9181740 DOI: 10.3390/ijms23116089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a toxic, aggregation-prone expansion of CAG repeats in the HTT gene with an age-dependent progression that leads to behavioral, cognitive and motor symptoms. Principally affecting the frontal cortex and the striatum, mHTT disrupts many cellular functions. In fact, increasing evidence shows that peripheral tissues are affected by neurodegenerative diseases. It establishes an active crosstalk between peripheral tissues and the brain in different neurodegenerative diseases. This review focuses on the current knowledge of peripheral tissue effects in HD animal and cell experimental models and identifies biomarkers and mechanisms involved or affected in the progression of the disease as new therapeutic or early diagnostic options. The particular changes in serum/plasma, blood cells such as lymphocytes, immune blood cells, the pancreas, the heart, the retina, the liver, the kidney and pericytes as a part of the blood-brain barrier are described. It is important to note that several changes in different mouse models of HD present differences between them and between the different ages analyzed. The understanding of the impact of peripheral organ inflammation in HD may open new avenues for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Laura Gómez-Jaramillo
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - Fátima Cano-Cano
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - María del Carmen González-Montelongo
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - Antonio Campos-Caro
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Área de Genética, Departamento de Biomedicina, Biotecnología y Salud Pública, Universidad de Cádiz, 11002 Cádiz, Spain
| | - Manuel Aguilar-Diosdado
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Departamento de Endocrinología y Nutrición, Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11002 Cádiz, Spain
| | - Ana I. Arroba
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Área de Genética, Departamento de Biomedicina, Biotecnología y Salud Pública, Universidad de Cádiz, 11002 Cádiz, Spain
| |
Collapse
|
18
|
Saba J, Couselo FL, Bruno J, Carniglia L, Durand D, Lasaga M, Caruso C. Neuroinflammation in Huntington's Disease: A Starring Role for Astrocyte and Microglia. Curr Neuropharmacol 2022; 20:1116-1143. [PMID: 34852742 PMCID: PMC9886821 DOI: 10.2174/1570159x19666211201094608] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/06/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative genetic disorder caused by a CAG repeat expansion in the huntingtin gene. HD causes motor, cognitive, and behavioral dysfunction. Since no existing treatment affects the course of this disease, new treatments are needed. Inflammation is frequently observed in HD patients before symptom onset. Neuroinflammation, characterized by the presence of reactive microglia, astrocytes and inflammatory factors within the brain, is also detected early. However, in comparison to other neurodegenerative diseases, the role of neuroinflammation in HD is much less known. Work has been dedicated to altered microglial and astrocytic functions in the context of HD, but less attention has been given to glial participation in neuroinflammation. This review describes evidence of inflammation in HD patients and animal models. It also discusses recent knowledge on neuroinflammation in HD, highlighting astrocyte and microglia involvement in the disease and considering anti-inflammatory therapeutic approaches.
Collapse
Affiliation(s)
- Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico López Couselo
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Bruno
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina,Address correspondence to this author at the Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155 Piso 10, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina, Tel: +54 11 5285 3380; E-mail:
| |
Collapse
|
19
|
Martí-Martínez S, Valor LM. A Glimpse of Molecular Biomarkers in Huntington's Disease. Int J Mol Sci 2022; 23:ijms23105411. [PMID: 35628221 PMCID: PMC9142992 DOI: 10.3390/ijms23105411] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder that is caused by an abnormal expansion of CAG repeats in the Huntingtin (HTT) gene. Although the main symptomatology is explained by alterations at the level of the central nervous system, predominantly affecting the basal ganglia, a peripheral component of the disease is being increasingly acknowledged. Therefore, the manifestation of the disease is complex and variable among CAG expansion carriers, introducing uncertainty in the appearance of specific signs, age of onset and severity of disease. The monogenic nature of the disorder allows a precise diagnosis, but the use of biomarkers with prognostic value is still needed to achieve clinical management of the patients in an individual manner. In addition, we need tools to evaluate the patient's response to potential therapeutic approaches. In this review, we provide a succinct summary of the most interesting molecular biomarkers that have been assessed in patients, mostly obtained from body fluids such as cerebrospinal fluid, peripheral blood and saliva.
Collapse
Affiliation(s)
- Silvia Martí-Martínez
- Servicio de Neurología, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain;
| | - Luis M. Valor
- Laboratorio de Apoyo a la Investigación, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
- Correspondence: ; Tel.: +34-965-913-988
| |
Collapse
|
20
|
Taheri F, Taghizadeh E, Navashenaq JG, Rezaee M, Gheibihayat SM. The role of efferocytosis in neuro-degenerative diseases. Neurol Sci 2022; 43:1593-1603. [PMID: 35059903 DOI: 10.1007/s10072-021-05835-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023]
Abstract
Efferocytosis has a critical role in maintaining tissues and organs' homeostasis by removing apoptotic cells. It is essential for human health, and disturbances in efferocytosis may result indifferent illnesses. In case of inadequate clearance of the dead cells, the content in the cells would be released. In fact, it induces some damages to the tissue and leads to the prolonged inflammation, so unsuitable phagocytosis of the apoptotic cells is involved in occurrence as well as expansion of numerous human chronic inflammatory diseases. Studies have shown age dependence of the neuro-degenerative diseases, which are largely due to the neuro-inflammation and the loss of neurons and thus cause the brain's functional disorders. Efferocytosis is coupled to anti-inflammatory responses that contribute to the elimination of the dying neurons in neuro-degenerative diseases, so its disruption may make a risk factor in numerous human chronic inflammatory diseases such as multiple sclerosis, Alzheimer's disease, glioblastoma, and Rett syndrome. This study is a review of the efferocytosis molecular pathways and their role in neuro-degenerative diseases in order to discover a new treatment option to cure patients.
Collapse
Affiliation(s)
- Forough Taheri
- Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Eskandar Taghizadeh
- Department of Medical Genetic, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, Iran.
| |
Collapse
|
21
|
O'Regan GC, Farag SH, Casey CS, Wood-Kaczmar A, Pocock JM, Tabrizi SJ, Andre R. Human Huntington's disease pluripotent stem cell-derived microglia develop normally but are abnormally hyper-reactive and release elevated levels of reactive oxygen species. J Neuroinflammation 2021; 18:94. [PMID: 33874957 PMCID: PMC8054367 DOI: 10.1186/s12974-021-02147-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/01/2021] [Indexed: 01/13/2023] Open
Abstract
Background Neuroinflammation may contribute to the pathogenesis of Huntington’s disease, given evidence of activated microglia and elevated levels of inflammatory molecules in disease gene carriers, even those many years from symptom onset. We have shown previously that monocytes from Huntington’s disease patients are hyper-reactive to stimulation in a manner dependent on their autonomous expression of the disease-causing mutant HTT protein. To date, however, whether human microglia are similarly hyper-responsive in a cell-autonomous manner has not been determined. Methods Microglial-like cells were derived from human pluripotent stem cells (PSCs) expressing mutant HTT containing varying polyglutamine lengths. These included lines that are otherwise isogenic, such that any observed differences can be attributed with certainty to the disease mutation itself. Analyses by quantitative PCR and immunofluorescence microscopy respectively of key genes and protein markers were undertaken to determine whether Huntington’s disease PSCs differentiated normally to a microglial fate. The resultant cultures and their supernatants were then assessed by various biochemical assays and multiplex ELISAs for viability and responses to stimulation, including the release of pro-inflammatory cytokines and reactive oxygen species. Conditioned media were applied to PSC-derived striatal neurons, and vice versa, to determine the effects that the secretomes of each cell type might have on the other. Results Human PSCs generated microglia successfully irrespective of the expression of mutant HTT. These cells, however, were hyper-reactive to stimulation in the production of pro-inflammatory cytokines such as IL-6 and TNFα. They also released elevated levels of reactive oxygen species that have neurotoxic potential. Accompanying such phenotypes, human Huntington’s disease PSC-derived microglia showed increased levels of apoptosis and were more susceptible to exogenous stress. Such stress appeared to be induced by supernatants from human PSC-derived striatal neurons expressing mutant HTT with a long polyglutamine tract. Conclusions These studies show, for the first time, that human Huntington’s disease PSC-derived microglia are hyper-reactive due to their autonomous expression of mutant HTT. This provides a cellular basis for the contribution that neuroinflammation might make to Huntington’s disease pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02147-6.
Collapse
Affiliation(s)
- Grace C O'Regan
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK
| | - Sahar H Farag
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK
| | - Caroline S Casey
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK
| | - Alison Wood-Kaczmar
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, WC1N 1PJ, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK.
| | - Ralph Andre
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, WC1N 3BG, London, UK.
| |
Collapse
|
22
|
Bondulich MK, Fan Y, Song Y, Giorgini F, Bates GP. Ablation of kynurenine 3-monooxygenase rescues plasma inflammatory cytokine levels in the R6/2 mouse model of Huntington's disease. Sci Rep 2021; 11:5484. [PMID: 33750843 PMCID: PMC7943810 DOI: 10.1038/s41598-021-84858-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/18/2021] [Indexed: 12/31/2022] Open
Abstract
Kynurenine 3-monooxygenase (KMO) regulates the levels of neuroactive metabolites in the kynurenine pathway (KP), dysregulation of which is associated with Huntington's disease (HD) pathogenesis. KMO inhibition leads to increased levels of neuroprotective relative to neurotoxic metabolites, and has been found to ameliorate disease-relevant phenotypes in several HD models. Here, we crossed KMO knockout mice to R6/2 HD mice to examine the effect of KMO depletion in the brain and periphery. KP genes were dysregulated in peripheral tissues from R6/2 mice and KMO ablation normalised levels of a subset of these. KP metabolites were also assessed, and KMO depletion led to increased levels of neuroprotective kynurenic acid in brain and periphery, and dramatically reduced neurotoxic 3-hydroxykunurenine levels in striatum and cortex. Notably, the increased levels of pro-inflammatory cytokines TNFa, IL1β, IL4 and IL6 found in R6/2 plasma were normalised upon KMO deletion. Despite these improvements in KP dysregulation and peripheral inflammation, KMO ablation had no effect upon several behavioural phenotypes. Therefore, although genetic inhibition of KMO in R6/2 mice modulates several metabolic and inflammatory parameters, these do not translate to improvements in primary disease indicators-observations which will likely be relevant for other interventions targeted at peripheral inflammation in HD.
Collapse
Affiliation(s)
- Marie Katrin Bondulich
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Yilan Fan
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Yeojin Song
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK.
| | - Gillian P Bates
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute At UCL, Queen Square Institute of Neurology, UCL, Queen Square, WC1N 3BG, UK
| |
Collapse
|
23
|
Przybyl L, Wozna-Wysocka M, Kozlowska E, Fiszer A. What, When and How to Measure-Peripheral Biomarkers in Therapy of Huntington's Disease. Int J Mol Sci 2021; 22:ijms22041561. [PMID: 33557131 PMCID: PMC7913877 DOI: 10.3390/ijms22041561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Among the main challenges in further advancing therapeutic strategies for Huntington’s disease (HD) is the development of biomarkers which must be applied to assess the efficiency of the treatment. HD is a dreadful neurodegenerative disorder which has its source of pathogenesis in the central nervous system (CNS) but is reflected by symptoms in the periphery. Visible symptoms include motor deficits and slight changes in peripheral tissues, which can be used as hallmarks for prognosis of the course of HD, e.g., the onset of the disease symptoms. Knowing how the pathology develops in the context of whole organisms is crucial for the development of therapy which would be the most beneficial for patients, as well as for proposing appropriate biomarkers to monitor disease progression and/or efficiency of treatment. We focus here on molecular peripheral biomarkers which could be used as a measurable outcome of potential therapy. We present and discuss a list of wet biomarkers which have been proposed in recent years to measure pre- and postsymptomatic HD. Interestingly, investigation of peripheral biomarkers in HD can unravel new aspects of the disease pathogenesis. This especially refers to inflammatory proteins or specific immune cells which attract scientific attention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Lukasz Przybyl
- Laboratory of Mammalian Model Organisms, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland
- Correspondence: (L.P.); (A.F.)
| | - Magdalena Wozna-Wysocka
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (M.W.-W.); (E.K.)
| | - Emilia Kozlowska
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (M.W.-W.); (E.K.)
| | - Agnieszka Fiszer
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (M.W.-W.); (E.K.)
- Correspondence: (L.P.); (A.F.)
| |
Collapse
|
24
|
O'Regan GC, Farag SH, Ostroff GR, Tabrizi SJ, Andre R. Wild-type huntingtin regulates human macrophage function. Sci Rep 2020; 10:17269. [PMID: 33057179 PMCID: PMC7560844 DOI: 10.1038/s41598-020-74042-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023] Open
Abstract
The huntingtin (HTT) protein in its mutant form is the cause of the inherited neurodegenerative disorder, Huntington's disease. Beyond its effects in the central nervous system, disease-associated mutant HTT causes aberrant phenotypes in myeloid-lineage innate immune system cells, namely monocytes and macrophages. Whether the wild-type form of the protein, however, has a role in normal human macrophage function has not been determined. Here, the effects of lowering the expression of wild-type (wt)HTT on the function of primary monocyte-derived macrophages from healthy, non-disease human subjects were examined. This demonstrated a previously undescribed role for wtHTT in maintaining normal macrophage health and function. Lowered wtHTT expression was associated, for instance, with a diminished release of induced cytokines, elevated phagocytosis and increased vulnerability to cellular stress. These may well occur by mechanisms different to that associated with the mutant form of the protein, given an absence of any effect on the intracellular signalling pathway predominantly associated with macrophage dysfunction in Huntington's disease.
Collapse
Affiliation(s)
- Grace C O'Regan
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Sahar H Farag
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Gary R Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Two Biotech, Suite 113, Worcester, MA, 01605, USA
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK. .,UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
| | - Ralph Andre
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
| |
Collapse
|
25
|
Scheiblich H, Trombly M, Ramirez A, Heneka MT. Neuroimmune Connections in Aging and Neurodegenerative Diseases. Trends Immunol 2020; 41:300-312. [DOI: 10.1016/j.it.2020.02.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 11/26/2022]
|
26
|
Lin YH, Maaroufi HO, Ibrahim E, Kucerova L, Zurovec M. Expression of Human Mutant Huntingtin Protein in Drosophila Hemocytes Impairs Immune Responses. Front Immunol 2019; 10:2405. [PMID: 31681295 PMCID: PMC6805700 DOI: 10.3389/fimmu.2019.02405] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/25/2019] [Indexed: 01/30/2023] Open
Abstract
The pathogenic effect of mutant HTT (mHTT) which causes Huntington disease (HD) are not restricted to nervous system. Such phenotypes include aberrant immune responses observed in the HD models. However, it is still unclear how this immune dysregulation influences the innate immune response against pathogenic infection. In the present study, we used transgenic Drosophila melanogaster expressing mutant HTT protein (mHTT) with hemocyte-specific drivers and examined the immune responses and hemocyte function. We found that mHTT expression in the hemocytes did not affect fly viability, but the numbers of circulating hemocytes were significantly decreased. Consequently, we observed that the expression of mHTT in the hemocytes compromised the immune responses including clot formation and encapsulation which lead to the increased susceptibility to entomopathogenic nematode and parasitoid wasp infections. In addition, mHTT expression in Drosophila macrophage-like S2 cells in vitro reduced ATP levels, phagocytic activity and the induction of antimicrobial peptides. Further effects observed in mHTT-expressing cells included the altered production of cytokines and activation of JAK/STAT signaling. The present study shows that the expression of mHTT in Drosophila hemocytes causes deficient cellular and humoral immune responses against invading pathogens. Our findings provide the insight into the pathogenic effects of mHTT in the immune cells.
Collapse
Affiliation(s)
- Yu-Hsien Lin
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Ceske Budejovice, Czechia.,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Houda Ouns Maaroufi
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Ceske Budejovice, Czechia.,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Emad Ibrahim
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Ceske Budejovice, Czechia.,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Lucie Kucerova
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Ceske Budejovice, Czechia
| | - Michal Zurovec
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Ceske Budejovice, Czechia.,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| |
Collapse
|
27
|
Valadão PAC, Oliveira BDS, Joviano-Santos JV, Vieira ÉLM, Rocha NP, Teixeira AL, Guatimosim C, de Miranda AS. Inflammatory changes in peripheral organs in the BACHD murine model of Huntington's disease. Life Sci 2019; 232:116653. [DOI: 10.1016/j.lfs.2019.116653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
|
28
|
Pido-Lopez J, Tanudjojo B, Farag S, Bondulich MK, Andre R, Tabrizi SJ, Bates GP. Inhibition of tumour necrosis factor alpha in the R6/2 mouse model of Huntington's disease by etanercept treatment. Sci Rep 2019; 9:7202. [PMID: 31076648 PMCID: PMC6510744 DOI: 10.1038/s41598-019-43627-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the expansion of the CAG repeat in exon 1 of the huntingtin (HTT) gene, which results in a mutant protein with an extended polyglutamine tract. Inflammation occurs in both the brain and the periphery of HD patients and mouse models, with increases in brain and/or plasma levels of neurotoxic TNFα and several other proinflammatory cytokines. TNFα promotes the generation of many of these cytokines, such as IL6, which raises the possibility that TNFα is central to the inflammatory milieu associated with HD. A number of mouse studies have reported that the suppression of chronic immune activation during HD has beneficial consequences. Here, we investigated whether TNFα contributes to the peripheral inflammation that occurs in the R6/2 mouse model, and whether the in vivo blockade of TNFα, via etanercept treatment, can modify disease progression. We found that etanercept treatment normalised the elevated plasma levels of some cytokines. This did not modify the progression of certain behavioural measures, but slightly ameliorated brain weight loss, possibly related to a reduction in the elevated striatal level of soluble TNFα.
Collapse
Affiliation(s)
- Jeffrey Pido-Lopez
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
| | - Benedict Tanudjojo
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.,Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Sahar Farag
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Marie-Katrin Bondulich
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ralph Andre
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Gillian P Bates
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
| |
Collapse
|
29
|
Askeland G, Rodinova M, Štufková H, Dosoudilova Z, Baxa M, Smatlikova P, Bohuslavova B, Klempir J, Nguyen TD, Kuśnierczyk A, Bjørås M, Klungland A, Hansikova H, Ellederova Z, Eide L. A transgenic minipig model of Huntington's disease shows early signs of behavioral and molecular pathologies. Dis Model Mech 2018; 11:dmm.035949. [PMID: 30254085 PMCID: PMC6215428 DOI: 10.1242/dmm.035949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Huntington's disease (HD) is a monogenic, progressive, neurodegenerative disorder with currently no available treatment. The Libechov transgenic minipig model for HD (TgHD) displays neuroanatomical similarities to humans and exhibits slow disease progression, and is therefore more powerful than available mouse models for the development of therapy. The phenotypic characterization of this model is still ongoing, and it is essential to validate biomarkers to monitor disease progression and intervention. In this study, the behavioral phenotype (cognitive, motor and behavior) of the TgHD model was assessed, along with biomarkers for mitochondrial capacity, oxidative stress, DNA integrity and DNA repair at different ages (24, 36 and 48 months), and compared with age-matched controls. The TgHD minipigs showed progressive accumulation of the mutant huntingtin (mHTT) fragment in brain tissue and exhibited locomotor functional decline at 48 months. Interestingly, this neuropathology progressed without any significant age-dependent changes in any of the other biomarkers assessed. Rather, we observed genotype-specific effects on mitochondrial DNA (mtDNA) damage, mtDNA copy number, 8-oxoguanine DNA glycosylase activity and global level of the epigenetic marker 5-methylcytosine that we believe is indicative of a metabolic alteration that manifests in progressive neuropathology. Peripheral blood mononuclear cells (PBMCs) were relatively spared in the TgHD minipig, probably due to the lack of detectable mHTT. Our data demonstrate that neuropathology in the TgHD model has an age of onset of 48 months, and that oxidative damage and electron transport chain impairment represent later states of the disease that are not optimal for assessing interventions. This article has an associated First Person interview with the first author of the paper. Summary: Here, we show that a minipig model of Huntington's disease mimics human neurodegeneration and holds promise for future intervention studies. However, minipig peripheral blood mononuclear cells express no detectable mutant huntingtin, eliminating their use as monitoring tools.
Collapse
Affiliation(s)
- Georgina Askeland
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway.,Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Marie Rodinova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12808, Czech Republic
| | - Hana Štufková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12808, Czech Republic
| | - Zaneta Dosoudilova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12808, Czech Republic
| | - Monika Baxa
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12808, Czech Republic
| | - Petra Smatlikova
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov 27721, Czech Republic
| | - Bozena Bohuslavova
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov 27721, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 12843, Czech Republic
| | - Jiri Klempir
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12821, Czech Republic
| | - The Duong Nguyen
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov 27721, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 12843, Czech Republic
| | - Anna Kuśnierczyk
- Proteomics and Metabolomics Core Facility, PROMEC, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Magnar Bjørås
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway.,Proteomics and Metabolomics Core Facility, PROMEC, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arne Klungland
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Hana Hansikova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague 12808, Czech Republic
| | - Zdenka Ellederova
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov 27721, Czech Republic
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
30
|
In vivo neutralization of the protagonist role of macrophages during the chronic inflammatory stage of Huntington's disease. Sci Rep 2018; 8:11447. [PMID: 30061661 PMCID: PMC6065433 DOI: 10.1038/s41598-018-29792-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases, characterised by the progressive and selective neuronal death in the central nervous system, are frequently accompanied by an activated immune system. In Huntington’s disease (HD), clinical and animal studies show evidence of immune activity, along with hyper-reactive monocyte/macrophage responses, while application of immunosuppressive regimens have imparted beneficial effects to HD mice. These findings suggest a contributory role of the immune system in HD pathology, with immune-based interventions offering a potential therapeutic strategy. Herein, we show that peripheral and CNS immune system activity increased with disease progression in HD mouse models and defined the phenotype of the immune response. Additionally, the depletion of monocytes and macrophages in vivo, via clodronate liposome treatment, revealed a major contributory role of these innate immune cells to the chronic inflammatory milieu observed during the course of the disease. This suggests that peripheral immunomodulatory strategies targeting monocytes and macrophages could be relevant for HD.
Collapse
|
31
|
Hensman Moss DJ, Robertson N, Farmer R, Scahill RI, Haider S, Tessari MA, Flynn G, Fischer DF, Wild EJ, Macdonald D, Tabrizi SJ. Quantification of huntingtin protein species in Huntington's disease patient leukocytes using optimised electrochemiluminescence immunoassays. PLoS One 2017; 12:e0189891. [PMID: 29272284 PMCID: PMC5741241 DOI: 10.1371/journal.pone.0189891] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/01/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is an autosomal dominant neurodegenerative condition caused by an expanded CAG repeat in the gene encoding huntingtin (HTT). Optimizing peripheral quantification of huntingtin throughout the course of HD is valuable not only to illuminate the natural history and pathogenesis of disease, but also to detect peripheral effects of drugs in clinical trial. RATIONALE We previously demonstrated that mutant HTT (mHTT) was significantly elevated in purified HD patient leukocytes compared with controls and that these levels track disease progression. Our present study investigates whether the same result can be achieved with a simpler and more scalable collection technique that is more suitable for clinical trials. METHODS We collected whole blood at 133 patient visits in two sample sets and generated peripheral blood mononuclear cells (PBMCs). Levels of mHTT, as well as N-, and C-terminal and mid-region huntingtin were measured in the PBMCs using ELISA-based Meso Scale Discovery (MSD) electrochemiluminescence immunoassay platforms, and we evaluated the relationship between different HTT species, disease stage, and brain atrophy on magnetic resonance imaging. CONCLUSIONS The assays were sensitive and accurate. We confirm our previous findings that mHTT increases with advancing disease stage in patient PBMCs, this time using a simple collection protocol and scalable assay.
Collapse
Affiliation(s)
- Davina J. Hensman Moss
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Nicola Robertson
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Ruth Farmer
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Rachael I. Scahill
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Salman Haider
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | | | | | | | - Edward J. Wild
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Douglas Macdonald
- CHDI Management/CHDI Foundation, Los Angeles, California, United States of America
| | - Sarah J. Tabrizi
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
32
|
Park M, Min B, Jeon K, Cho S, Park JS, Kim J, Jeon J, Song J, Kim S, Jeong S, Seo H, Kang YK. Age-associated chromatin relaxation is enhanced in Huntington's disease mice. Aging (Albany NY) 2017; 9:803-822. [PMID: 28288000 PMCID: PMC5391233 DOI: 10.18632/aging.101193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 02/26/2017] [Indexed: 12/13/2022]
Abstract
Expansion of polyglutamine stretch in the huntingtin (HTT) protein is a major cause of Huntington's disease (HD). The polyglutamine part in HTT interacts with various proteins implicated in epigenetic regulation of genes, suggesting that mutant HTT may disturb the integrity of the epigenetic system. Here, we used a PCRseq-based method to examine expression profile of 395 exonic segments from 260 “epi-driver” genes in splenic T lymphocytes from aged HD mice. We identified 67 exonic segments differentially expressed between young and aged HD mice, most of them upregulated in the aged. Polycomb-repressive complex (PRC)-regulated genes (PRGs) were markedly upregulated in aged HD mice, consistent with downregulation of PRC genes. Epi-driver gene categories of lysine-methylation, lysine-demethylation, arginine-methylation, and PRG showed differential age-associated changes between HD and control. Analyzing the pattern of change in epi-driver gene expressions hinted at an enhanced shift in HD chromatin to a more accessible state with age, which was experimentally demonstrated by DNase-I-hypersensitivity sequencing showing increased chromatin accessibility in HD cells compared to control. We suggest the global change can potentially relieve chromatin-induced repression of many genes, and the unintended expressions of some detrimental proteins could alter T cell function to a greater degree in aged HD mice.
Collapse
Affiliation(s)
- Myungsun Park
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea
| | - Byungkuk Min
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea
| | - Kyuheum Jeon
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea.,Department of Functional Genomics, University of Science and Technology (UST), Yuseong-gu, Daejeon, 305-350, South Korea
| | - Sunwha Cho
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea
| | - Jung Sun Park
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea.,Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea
| | - Jisun Kim
- Department of Molecular and Life Sciences, Hanyang University, Sangnok-gu, Ansan, Gyeonggi-do, 15588, South Korea
| | - Jeha Jeon
- Department of Molecular and Life Sciences, Hanyang University, Sangnok-gu, Ansan, Gyeonggi-do, 15588, South Korea
| | - Jinhoi Song
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea
| | - Seokho Kim
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea
| | - Sangkyun Jeong
- Mibyeong Research Center, Korea Institute of Oriental Medicine (KIOM), Yuseong-gu, Daejeon, 305-811, South Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Sangnok-gu, Ansan, Gyeonggi-do, 15588, South Korea
| | - Yong-Kook Kang
- Development and Differentiation Research Center, KRIBB, Yuseong-gu, Daejeon, 305-806, South Korea.,Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea.,Department of Functional Genomics, University of Science and Technology (UST), Yuseong-gu, Daejeon, 305-350, South Korea
| |
Collapse
|
33
|
Al-Gharaibeh A, Culver R, Stewart AN, Srinageshwar B, Spelde K, Frollo L, Kolli N, Story D, Paladugu L, Anwar S, Crane A, Wyse R, Maiti P, Dunbar GL, Rossignol J. Induced Pluripotent Stem Cell-Derived Neural Stem Cell Transplantations Reduced Behavioral Deficits and Ameliorated Neuropathological Changes in YAC128 Mouse Model of Huntington's Disease. Front Neurosci 2017; 11:628. [PMID: 29209158 PMCID: PMC5701605 DOI: 10.3389/fnins.2017.00628] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023] Open
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disorder characterized by neuronal loss and motor dysfunction. Although there is no effective treatment, stem cell transplantation offers a promising therapeutic strategy, but the safety and efficacy of this approach needs to be optimized. The purpose of this study was to test the potential of intra-striatal transplantation of induced pluripotent stem cell-derived neural stem cells (iPS-NSCs) for treating HD. For this purpose, we developed mouse adenovirus-generated iPSCs, differentiated them into neural stem cells in vitro, labeled them with Hoechst, and transplanted them bilaterally into striata of 10-month old wild type (WT) and HD YAC128 mice. We assessed the efficiency of these transplanted iPS-NSCs to reduce motor deficits in YAC128 mice by testing them on an accelerating rotarod task at 1 day prior to transplantation, and then weekly for 10 weeks. Our results showed an amelioration of locomotor deficits in YAC128 mice that received iPS-NSC transplantations. Following testing, the mice were sacrificed, and their brains were analyzed using immunohistochemistry and Western blot (WB). The results from our histological examinations revealed no signs of tumors and evidence that many iPS-NSCs survived and differentiated into region-specific neurons (medium spiny neurons) in both WT and HD mice, as confirmed by co-labeling of Hoechst-labeled transplanted cells with NeuN and DARPP-32. Also, counts of Hoechst-labeled cells revealed that a higher proportion were co-labeled with DARPP-32 and NeuN in HD-, compared to WT- mice, suggesting a dissimilar differentiation pattern in HD mice. Whereas significant decreases were found in counts of NeuN- and DARPP-32-labeled cells, and for neuronal density measures in striata of HD vehicle controls, such decrements were not observed in the iPS-NSCs-transplanted-HD mice. WB analysis showed increase of BDNF and TrkB levels in striata of transplanted HD mice compared to HD vehicle controls. Collectively, our data suggest that iPS-NSCs may provide an effective option for neuronal replacement therapy in HD.
Collapse
Affiliation(s)
- Abeer Al-Gharaibeh
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Rebecca Culver
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Andrew N Stewart
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Bhairavi Srinageshwar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Kristin Spelde
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Laura Frollo
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Nivya Kolli
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Darren Story
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States
| | - Leela Paladugu
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Sarah Anwar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Andrew Crane
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Robert Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, United States
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, United States
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mt Pleasant, MI, United States
| |
Collapse
|
34
|
Bondulich MK, Jolinon N, Osborne GF, Smith EJ, Rattray I, Neueder A, Sathasivam K, Ahmed M, Ali N, Benjamin AC, Chang X, Dick JRT, Ellis M, Franklin SA, Goodwin D, Inuabasi L, Lazell H, Lehar A, Richard-Londt A, Rosinski J, Smith DL, Wood T, Tabrizi SJ, Brandner S, Greensmith L, Howland D, Munoz-Sanjuan I, Lee SJ, Bates GP. Myostatin inhibition prevents skeletal muscle pathophysiology in Huntington's disease mice. Sci Rep 2017; 7:14275. [PMID: 29079832 PMCID: PMC5660167 DOI: 10.1038/s41598-017-14290-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/06/2017] [Indexed: 11/09/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder of which skeletal muscle atrophy is a common feature, and multiple lines of evidence support a muscle-based pathophysiology in HD mouse models. Inhibition of myostatin signaling increases muscle mass, and therapeutic approaches based on this are in clinical development. We have used a soluble ActRIIB decoy receptor (ACVR2B/Fc) to test the effects of myostatin/activin A inhibition in the R6/2 mouse model of HD. Weekly administration from 5 to 11 weeks of age prevented body weight loss, skeletal muscle atrophy, muscle weakness, contractile abnormalities, the loss of functional motor units in EDL muscles and delayed end-stage disease. Inhibition of myostatin/activin A signaling activated transcriptional profiles to increase muscle mass in wild type and R6/2 mice but did little to modulate the extensive Huntington's disease-associated transcriptional dysregulation, consistent with treatment having little impact on HTT aggregation levels. Modalities that inhibit myostatin signaling are currently in clinical trials for a variety of indications, the outcomes of which will present the opportunity to assess the potential benefits of targeting this pathway in HD patients.
Collapse
Affiliation(s)
- Marie K Bondulich
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Nelly Jolinon
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Georgina F Osborne
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Edward J Smith
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Ivan Rattray
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Andreas Neueder
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Kirupa Sathasivam
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Mhoriam Ahmed
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Nadira Ali
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Agnesska C Benjamin
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Xiaoli Chang
- Department Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - James R T Dick
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Matthew Ellis
- Division of Neuropathology, UCL Institute of Neurology, London, WC1N 3BG, UK
- Department of Neurodegenerative disease, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Sophie A Franklin
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Daniel Goodwin
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Linda Inuabasi
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Hayley Lazell
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Adam Lehar
- Department Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Angela Richard-Londt
- Division of Neuropathology, UCL Institute of Neurology, London, WC1N 3BG, UK
- Department of Neurodegenerative disease, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Jim Rosinski
- CHDI Management/CHDI Foundation Inc, New York, NY, 10001, USA
| | - Donna L Smith
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Tobias Wood
- Department of Neuroimaging, King's College London, Institute of Psychiatry, London, SE5 8AF, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
- Department of Neurodegenerative disease, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Sebastian Brandner
- Division of Neuropathology, UCL Institute of Neurology, London, WC1N 3BG, UK
- Department of Neurodegenerative disease, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - David Howland
- CHDI Management/CHDI Foundation Inc, New York, NY, 10001, USA
| | | | - Se-Jin Lee
- Department Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Gillian P Bates
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, WC1N 3BG, UK.
- Department Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK.
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
35
|
Sjögren M, Duarte AI, McCourt AC, Shcherbina L, Wierup N, Björkqvist M. Ghrelin rescues skeletal muscle catabolic profile in the R6/2 mouse model of Huntington's disease. Sci Rep 2017; 7:13896. [PMID: 29066728 PMCID: PMC5654969 DOI: 10.1038/s41598-017-13713-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests altered energy metabolism as a key feature in Huntington’s disease (HD) pathology. Hyper-catabolism, including weight loss and muscle atrophy, is seen in HD patients and HD mouse models. Metabolic hormones are key players, not only in energy metabolism, but also in neurodegenerative processes. Ghrelin, a gut peptide-hormone, plays an important role in regulating energy metabolism, stimulating appetite, and affects brain function and increases neuronal survival. The R6/2 mouse model of HD has previously been shown to exhibit progressive weight loss, dysregulated glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using ghrelin administration, with the primary aim to delay weight loss and reduce muscle atrophy. We also evaluated glucose metabolism and behaviour. We here demonstrate that ghrelin administration (subcutaneous 150 μg/kg daily injections) for 4 weeks, reversed the catabolic gene expression profile (increased expression of Caspase 8, Traf-5 and Creb1) seen in R6/2 mouse skeletal muscle. Skeletal muscle morphology was also improved with ghrelin, and importantly, ghrelin administration normalized behavioural deficits in R6/2 mice. Taken together, our findings encourage further studies targeting metabolism in HD.
Collapse
Affiliation(s)
- Marie Sjögren
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Brain Disease Biomarker Unit, Lund University, Lund, Sweden.
| | - Ana I Duarte
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Brain Disease Biomarker Unit, Lund University, Lund, Sweden.,CNC - Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - Andrew C McCourt
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Brain Disease Biomarker Unit, Lund University, Lund, Sweden
| | - Liliya Shcherbina
- Lund University Diabetes Centre, Neuroendocrine Cell Biology, Department of Clinical Sciences in Malmö, Clinical research center, Lund University, Malmö, Sweden
| | - Nils Wierup
- Lund University Diabetes Centre, Neuroendocrine Cell Biology, Department of Clinical Sciences in Malmö, Clinical research center, Lund University, Malmö, Sweden
| | - Maria Björkqvist
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Brain Disease Biomarker Unit, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Miller JRC, Pfister EL, Liu W, Andre R, Träger U, Kennington LA, Lo K, Dijkstra S, Macdonald D, Ostroff G, Aronin N, Tabrizi SJ. Allele-Selective Suppression of Mutant Huntingtin in Primary Human Blood Cells. Sci Rep 2017; 7:46740. [PMID: 28436437 PMCID: PMC5402279 DOI: 10.1038/srep46740] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/27/2017] [Indexed: 01/31/2023] Open
Abstract
Post-transcriptional gene silencing is a promising therapy for the monogenic, autosomal dominant, Huntington's disease (HD). However, wild-type huntingtin (HTT) has important cellular functions, so the ideal strategy would selectively lower mutant HTT while sparing wild-type. HD patients were genotyped for heterozygosity at three SNP sites, before phasing each SNP allele to wild-type or mutant HTT. Primary ex vivo myeloid cells were isolated from heterozygous patients and transfected with SNP-targeted siRNA, using glucan particles taken up by phagocytosis. Highly selective mRNA knockdown was achieved when targeting each allele of rs362331 in exon 50 of the HTT transcript; this selectivity was also present on protein studies. However, similar selectivity was not observed when targeting rs362273 or rs362307. Furthermore, HD myeloid cells are hyper-reactive compared to control. Allele-selective suppression of either wild-type or mutant HTT produced a significant, equivalent reduction in the cytokine response of HD myeloid cells to LPS, suggesting that wild-type HTT has a novel immune function. We demonstrate a sequential therapeutic process comprising genotyping and mutant HTT-linkage of SNPs, followed by personalised allele-selective suppression in a small patient cohort. We further show that allele-selectivity in ex vivo patient cells is highly SNP-dependent, with implications for clinical trial target selection.
Collapse
Affiliation(s)
- James R. C. Miller
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Edith L. Pfister
- RNA Therapeutics Institute and Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Wanzhao Liu
- RNA Therapeutics Institute and Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ralph Andre
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ulrike Träger
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Lori A. Kennington
- RNA Therapeutics Institute and Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | - Gary Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Neil Aronin
- RNA Therapeutics Institute and Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sarah J. Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| |
Collapse
|
37
|
Hensman Moss DJ, Flower MD, Lo KK, Miller JRC, van Ommen GJB, ’t Hoen PAC, Stone TC, Guinee A, Langbehn DR, Jones L, Plagnol V, van Roon-Mom WMC, Holmans P, Tabrizi SJ. Huntington's disease blood and brain show a common gene expression pattern and share an immune signature with Alzheimer's disease. Sci Rep 2017; 7:44849. [PMID: 28322270 PMCID: PMC5359597 DOI: 10.1038/srep44849] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/14/2017] [Indexed: 12/25/2022] Open
Abstract
There is widespread transcriptional dysregulation in Huntington's disease (HD) brain, but analysis is inevitably limited by advanced disease and postmortem changes. However, mutant HTT is ubiquitously expressed and acts systemically, meaning blood, which is readily available and contains cells that are dysfunctional in HD, could act as a surrogate for brain tissue. We conducted an RNA-Seq transcriptomic analysis using whole blood from two HD cohorts, and performed gene set enrichment analysis using public databases and weighted correlation network analysis modules from HD and control brain datasets. We identified dysregulated gene sets in blood that replicated in the independent cohorts, correlated with disease severity, corresponded to the most significantly dysregulated modules in the HD caudate, the most prominently affected brain region, and significantly overlapped with the transcriptional signature of HD myeloid cells. High-throughput sequencing technologies and use of gene sets likely surmounted the limitations of previously inconsistent HD blood expression studies. Our results suggest transcription is disrupted in peripheral cells in HD through mechanisms that parallel those in brain. Immune upregulation in HD overlapped with Alzheimer's disease, suggesting a common pathogenic mechanism involving macrophage phagocytosis and microglial synaptic pruning, and raises the potential for shared therapeutic approaches.
Collapse
Affiliation(s)
- Davina J. Hensman Moss
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London, WC1B 5EH, UK
| | - Michael D. Flower
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London, WC1B 5EH, UK
| | - Kitty K. Lo
- University College London Genetics Institute, University College London, London, WC1E 6BT, UK
| | - James R. C. Miller
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London, WC1B 5EH, UK
| | - Gert-Jan B. van Ommen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postzone S-4-P, The Netherlands
| | - Peter A. C. ’t Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postzone S-4-P, The Netherlands
| | - Timothy C. Stone
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, CF24 4HQ, UK
| | - Amelia Guinee
- Faculty of Education, University of Cambridge, CB2 8PQ, Cambridge UK
| | - Douglas R. Langbehn
- Departments of Psychiatry and Biostatistics, University of Iowa, IA 52242, USA
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, CF24 4HQ, UK
| | - Vincent Plagnol
- University College London Genetics Institute, University College London, London, WC1E 6BT, UK
| | | | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, CF24 4HQ, UK
| | - Sarah J. Tabrizi
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London, WC1B 5EH, UK
| |
Collapse
|
38
|
Wright DJ, Renoir T, Gray LJ, Hannan AJ. Huntington’s Disease: Pathogenic Mechanisms and Therapeutic Targets. ADVANCES IN NEUROBIOLOGY 2017; 15:93-128. [DOI: 10.1007/978-3-319-57193-5_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
39
|
Disatnik MH, Joshi AU, Saw NL, Shamloo M, Leavitt BR, Qi X, Mochly-Rosen D. Potential biomarkers to follow the progression and treatment response of Huntington's disease. J Exp Med 2016; 213:2655-2669. [PMID: 27821553 PMCID: PMC5110026 DOI: 10.1084/jem.20160776] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/16/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022] Open
Abstract
Disatnik et al. identify mitochondrial DNA levels, 8-OHdG, and inflammation factors as potential peripheral biomarkers to follow progression and treatment response of Huntington’s disease. Huntington’s disease (HD) is a rare genetic disease caused by expanded polyglutamine repeats in the huntingtin protein resulting in selective neuronal loss. Although genetic testing readily identifies those who will be affected, current pharmacological treatments do not prevent or slow down disease progression. A major challenge is the slow clinical progression and the inability to biopsy the affected tissue, the brain, making it difficult to design short and effective proof of concept clinical trials to assess treatment benefit. In this study, we focus on identifying peripheral biomarkers that correlate with the progression of the disease and treatment benefit. We recently developed an inhibitor of pathological mitochondrial fragmentation, P110, to inhibit neurotoxicity in HD. Changes in levels of mitochondrial DNA (mtDNA) and inflammation markers in plasma, a product of DNA oxidation in urine, mutant huntingtin aggregates, and 4-hydroxynonenal adducts in muscle and skin tissues were all noted in HD R6/2 mice relative to wild-type mice. Importantly, P110 treatment effectively reduced the levels of these biomarkers. Finally, abnormal levels of mtDNA were also found in plasma of HD patients relative to control subjects. Therefore, we identified several potential peripheral biomarkers as candidates to assess HD progression and the benefit of intervention for future clinical trials.
Collapse
Affiliation(s)
- Marie-Hélène Disatnik
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Amit U Joshi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Nay L Saw
- Behavioral and Functional Neuroscience Laboratory, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Mehrdad Shamloo
- Behavioral and Functional Neuroscience Laboratory, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
40
|
Huntingtons Disease Mice Infected with Toxoplasma gondii Demonstrate Early Kynurenine Pathway Activation, Altered CD8+ T-Cell Responses, and Premature Mortality. PLoS One 2016; 11:e0162404. [PMID: 27611938 PMCID: PMC5017698 DOI: 10.1371/journal.pone.0162404] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/10/2016] [Indexed: 12/03/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a polyglutamine-repeat expansion in the huntingtin protein. Activation of the kynurenine pathway of tryptophan degradation is implicated in the pathogenesis of HD. Indoleamine-2,3-dioxygenase (IDO) catalyzes the oxidation of tryptophan to kynurenine, the first step in this pathway. The prevalent, neuroinvasive protozoal pathogen Toxoplasma gondii (T. gondii) results in clinically silent life-long infection in immune-competent individuals. T. gondii infection results in activation of IDO which provides some protection against the parasite by depleting tryptophan which the parasite cannot synthesize. The kynurenine pathway may therefore represent a point of synergism between HD and T. gondii infection. We show here that IDO activity is elevated at least four-fold in frontal cortex and striata of non-infected N171-82Q HD mice at 14-weeks corresponding to early–advanced HD. T. gondii infection at 5 weeks resulted in elevation of cortical IDO activity in HD mice. HD-infected mice died significantly earlier than wild-type infected and HD control mice. Prior to death, infected HD mice demonstrated decreased CD8+ T-lymphocyte proliferation in brain and spleen compared to wild-type infected mice. We demonstrate for the first time that HD mice have an altered response to an infectious agent that is characterized by premature mortality, altered immune responses and early activation of IDO. Findings are relevant to understanding how T. gondii infection may interact with pathways mediating neurodegeneration in HD.
Collapse
|
41
|
Laquinimod rescues striatal, cortical and white matter pathology and results in modest behavioural improvements in the YAC128 model of Huntington disease. Sci Rep 2016; 6:31652. [PMID: 27528441 PMCID: PMC4985819 DOI: 10.1038/srep31652] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence supports a role for abnormal immune activation and inflammatory responses in Huntington disease (HD). In this study, we evaluated the therapeutic potential of laquinimod (1 and 10 mg/kg), a novel immunomodulatory agent shown to be protective in a number of neuroinflammatory conditions, in the YAC128 mouse model of HD. Treatment with laquinimod for 6 months rescued atrophy in the striatum, in certain cortical regions, and in the corpus callosum of YAC128 HD mice. Diffusion tensor imaging showed that white matter microstructural abnormalities in the posterior corpus callosum were improved following treatment with low dose (1 mg/kg) laquinimod, and were paralleled by reduced levels of interleukin-6 in the periphery of YAC128 HD mice. Functionally, treatment with laquinimod (1 and 10 mg/kg) led to modest improvements in motor function and in depressive-like behaviour. Taken together, these results suggest that laquinimod may improve some features of pathology in HD, and provides support for the role of immune activation in the pathogenesis of HD.
Collapse
|
42
|
Neuroimmunology of Huntington's Disease: Revisiting Evidence from Human Studies. Mediators Inflamm 2016; 2016:8653132. [PMID: 27578922 PMCID: PMC4992798 DOI: 10.1155/2016/8653132] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/12/2016] [Indexed: 12/11/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by selective loss of neurons in the striatum and cortex, which leads to progressive motor dysfunction, cognitive decline, and psychiatric disorders. Although the cause of HD is well described—HD is a genetic disorder caused by a trinucleotide (CAG) repeat expansion in the gene encoding for huntingtin (HTT) on chromosome 4p16.3—the ultimate cause of neuronal death is still uncertain. Apart from impairment in systems for handling abnormal proteins, other metabolic pathways and mechanisms might contribute to neurodegeneration and progression of HD. Among these, inflammation seems to play a role in HD pathogenesis. The current review summarizes the available evidence about immune and/or inflammatory changes in HD. HD is associated with increased inflammatory mediators in both the central nervous system and periphery. Accordingly, there have been some attempts to slow HD progression targeting the immune system.
Collapse
|
43
|
Miller JRC, Lo KK, Andre R, Hensman Moss DJ, Träger U, Stone TC, Jones L, Holmans P, Plagnol V, Tabrizi SJ. RNA-Seq of Huntington's disease patient myeloid cells reveals innate transcriptional dysregulation associated with proinflammatory pathway activation. Hum Mol Genet 2016; 25:2893-2904. [PMID: 27170315 PMCID: PMC5181590 DOI: 10.1093/hmg/ddw142] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022] Open
Abstract
Innate immune activation beyond the central nervous system is emerging as a vital component of the pathogenesis of neurodegeneration. Huntington's disease (HD) is a fatal neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The systemic innate immune system is thought to act as a modifier of disease progression; however, the molecular mechanisms remain only partially understood. Here we use RNA-sequencing to perform whole transcriptome analysis of primary monocytes from thirty manifest HD patients and thirty-three control subjects, cultured with and without a proinflammatory stimulus. In contrast with previous studies that have required stimulation to elicit phenotypic abnormalities, we demonstrate significant transcriptional differences in HD monocytes in their basal, unstimulated state. This includes previously undetected increased resting expression of genes encoding numerous proinflammatory cytokines, such as IL6 Further pathway analysis revealed widespread resting enrichment of proinflammatory functional gene sets, while upstream regulator analysis coupled with Western blotting suggests that abnormal basal activation of the NFĸB pathway plays a key role in mediating these transcriptional changes. That HD myeloid cells have a proinflammatory phenotype in the absence of stimulation is consistent with a priming effect of mutant huntingtin, whereby basal dysfunction leads to an exaggerated inflammatory response once a stimulus is encountered. These data advance our understanding of mutant huntingtin pathogenesis, establish resting myeloid cells as a key source of HD immune dysfunction, and further demonstrate the importance of systemic immunity in the potential treatment of HD and the wider study of neurodegeneration.
Collapse
Affiliation(s)
- James R C Miller
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kitty K Lo
- UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Ralph Andre
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Davina J Hensman Moss
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ulrike Träger
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Timothy C Stone
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Vincent Plagnol
- UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| |
Collapse
|
44
|
Valenza M, Chen JY, Di Paolo E, Ruozi B, Belletti D, Ferrari Bardile C, Leoni V, Caccia C, Brilli E, Di Donato S, Boido MM, Vercelli A, Vandelli MA, Forni F, Cepeda C, Levine MS, Tosi G, Cattaneo E. Cholesterol-loaded nanoparticles ameliorate synaptic and cognitive function in Huntington's disease mice. EMBO Mol Med 2016; 7:1547-64. [PMID: 26589247 PMCID: PMC4693506 DOI: 10.15252/emmm.201505413] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Brain cholesterol biosynthesis and cholesterol levels are reduced in mouse models of Huntington's disease (HD), suggesting that locally synthesized, newly formed cholesterol is less available to neurons. This may be detrimental for neuronal function, especially given that locally synthesized cholesterol is implicated in synapse integrity and remodeling. Here, we used biodegradable and biocompatible polymeric nanoparticles (NPs) modified with glycopeptides (g7) and loaded with cholesterol (g7‐NPs‐Chol), which per se is not blood–brain barrier (BBB) permeable, to obtain high‐rate cholesterol delivery into the brain after intraperitoneal injection in HD mice. We report that g7‐NPs, in contrast to unmodified NPs, efficiently crossed the BBB and localized in glial and neuronal cells in different brain regions. We also found that repeated systemic delivery of g7‐NPs‐Chol rescued synaptic and cognitive dysfunction and partially improved global activity in HD mice. These results demonstrate that cholesterol supplementation to the HD brain reverses functional alterations associated with HD and highlight the potential of this new drug‐administration route to the diseased brain.
Collapse
Affiliation(s)
- Marta Valenza
- Department of BioSciences, Centre for Stem Cell Research Università degli Studi di Milano, Milan, Italy
| | - Jane Y Chen
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience Brain Research Institute David Geffen School of Medicine University of California Los Angeles, Los Angeles, CA, USA
| | - Eleonora Di Paolo
- Department of BioSciences, Centre for Stem Cell Research Università degli Studi di Milano, Milan, Italy
| | - Barbara Ruozi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Belletti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Costanza Ferrari Bardile
- Department of BioSciences, Centre for Stem Cell Research Università degli Studi di Milano, Milan, Italy
| | - Valerio Leoni
- Neurological Institute C. Besta, Milan, Italy Laboratory of Clinical Chemistry, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | | | - Elisa Brilli
- Department of BioSciences, Centre for Stem Cell Research Università degli Studi di Milano, Milan, Italy
| | | | - Marina M Boido
- Neuroscience Institute Cavalieri Ottolenghi Neuroscience Institute of Turin, Orbassano Turin, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi Neuroscience Institute of Turin, Orbassano Turin, Italy
| | - Maria A Vandelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Flavio Forni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience Brain Research Institute David Geffen School of Medicine University of California Los Angeles, Los Angeles, CA, USA
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience Brain Research Institute David Geffen School of Medicine University of California Los Angeles, Los Angeles, CA, USA
| | - Giovanni Tosi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Cattaneo
- Department of BioSciences, Centre for Stem Cell Research Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
45
|
DJ-1 deficiency attenuates expansion of liver progenitor cells through modulating the inflammatory and fibrogenic niches. Cell Death Dis 2016; 7:e2257. [PMID: 27277679 PMCID: PMC5143389 DOI: 10.1038/cddis.2016.161] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/27/2022]
Abstract
Our previous study suggested that DJ-1 has a critical role in initiating an inflammatory response, but its role in the liver progenitor cell (LPC) expansion, a process highly dependent on the inflammatory niche, remains elusive. The objective of this study is to determine the role of DJ-1 in LPC expansion. The correlation of DJ-1 expression with LPC markers was examined in the liver of patients with hepatitis B or hepatitis C virus (HBV and HCV, respectively) infection, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), nonalcoholic fatty liver disease (NAFLD), cirrhosis or hepatocellular carcinoma (HCC), respectively. The role of DJ-1 in LPC expansion and the formation of LPC-associated fibrosis and inflammation was examined in a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet-induced liver injury murine model. We also determined the ability of hepatic stellate cells (HSCs) in recruiting macrophages in DJ-1 knockout (KO) mice. The expression levels of DJ-1 were upregulated in the liver of HBV, HCV, PBC and PSC patients and DDC-fed mice. Additionally, DJ-1 expression was positively correlated with LPC proliferation in patients with liver injury and mice with DDC exposure. DJ-1 has no direct effect on LPC proliferation. Reduced activation of HSCs and collagen deposition were observed in DJ-1 KO mice. Furthermore, infiltrated CD11b+Gr-1low macrophages and pro-inflammatory factors (IL-6, TNF-α) were attenuated in DJ-1 KO mice. Mechanistically, we found that HSCs isolated from DJ-1 KO mice had decreased secretion of macrophage-mobilizing chemokines, such as CCL2 and CX3CL1, resulting in impaired macrophage infiltration. DJ-1 positively correlates with LPC expansion during liver injury. DJ-1 deficiency negatively regulates LPC proliferation by impairing the formation of LPC-associated fibrosis and inflammatory niches.
Collapse
|
46
|
Fan HC, Chi CS, Cheng SN, Lee HF, Tsai JD, Lin SZ, Harn HJ. Targeting New Candidate Genes by Small Molecules Approaching Neurodegenerative Diseases. Int J Mol Sci 2015; 17:E26. [PMID: 26712747 PMCID: PMC4730273 DOI: 10.3390/ijms17010026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/10/2015] [Accepted: 12/21/2015] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative diseases (NDs) are among the most feared of the disorders that afflict humankind for the lack of specific diagnostic tests and effective treatments. Understanding the molecular, cellular, biochemical changes of NDs may hold therapeutic promise against debilitating central nerve system (CNS) disorders. In the present review, we summarized the clinical presentations and biology backgrounds of NDs, including Parkinson's disease (PD), Huntington's disease (HD), and Alzheimer's disease (AD) and explored the role of molecular mechanisms, including dys-regulation of epigenetic control mechanisms, Ataxia-telangiectasia-mutated protein kinase (ATM), and neuroinflammation in the pathogenesis of NDs. Targeting these mechanisms may hold therapeutic promise against these devastating diseases.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tung's Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan.
| | - Ching-Shiang Chi
- Department of Pediatrics, Tung's Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan.
| | - Shin-Nan Cheng
- Department of Pediatrics, Tung's Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan.
| | - Hsiu-Fen Lee
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung 407, Taiwan.
| | - Jeng-Dau Tsai
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan.
| | - Shinn-Zong Lin
- Graduate Institute of Immunology, China Medical University, Taichung 404, Taiwan.
- Center for Neuropsychiatry, China Medical University and Hospital, Taichung 404, Taiwan.
- Department of Neurosurgery, China Medical University Beigang Hospital, Yunlin 651, Taiwan.
| | - Horng-Jyh Harn
- Department of Pathology, China Medical University and Hospital, Taichung 404, Taiwan.
| |
Collapse
|
47
|
Kerkis I, Haddad MS, Valverde CW, Glosman S. Neural and mesenchymal stem cells in animal models of Huntington's disease: past experiences and future challenges. Stem Cell Res Ther 2015; 6:232. [PMID: 26667114 PMCID: PMC4678723 DOI: 10.1186/s13287-015-0248-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD) is an inherited disease that causes progressive nerve cell degeneration. It is triggered by a mutation in the HTT gene that strongly influences functional abilities and usually results in movement, cognitive and psychiatric disorders. HD is incurable, although treatments are available to help manage symptoms and to delay the physical, mental and behavioral declines associated with the condition. Stem cells are the essential building blocks of life, and play a crucial role in the genesis and development of all higher organisms. Ablative surgical procedures and fetal tissue cell transplantation, which are still experimental, demonstrate low rates of recovery in HD patients. Due to neuronal cell death caused by accumulation of the mutated huntingtin (mHTT) protein, it is unlikely that such brain damage can be treated solely by drug-based therapies. Stem cell-based therapies are important in order to reconstruct damaged brain areas in HD patients. These therapies have a dual role: stem cell paracrine action, stimulating local cell survival, and brain tissue regeneration through the production of new neurons from the intrinsic and likely from donor stem cells. This review summarizes current knowledge on neural stem/progenitor cell and mesenchymal stem cell transplantation, which has been carried out in several animal models of HD, discussing cell distribution, survival and differentiation after transplantation, as well as functional recovery and anatomic improvements associated with these approaches. We also discuss the usefulness of this information for future preclinical and clinical studies in HD.
Collapse
Affiliation(s)
- Irina Kerkis
- Laboratório de Genética, Instituto Butantan, 1500 Av. Vital Brasil, São Paulo, 05503-900, Brazil.
| | - Monica Santoro Haddad
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 455 Av. Dr. Arnaldao, São Paulo, 01246903, Brazil
| | | | - Sabina Glosman
- SoluBest Ltd, Weizmann Science Park, POB 4053 18 Einstein Street, Ness Ziona, 74140, Israel
| |
Collapse
|
48
|
Miller JRC, Träger U, Andre R, Tabrizi SJ. Mutant Huntingtin Does Not Affect the Intrinsic Phenotype of Human Huntington's Disease T Lymphocytes. PLoS One 2015; 10:e0141793. [PMID: 26529236 PMCID: PMC4631523 DOI: 10.1371/journal.pone.0141793] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/13/2015] [Indexed: 11/28/2022] Open
Abstract
Huntington’s disease is a fatal neurodegenerative condition caused by a CAG repeat expansion in the huntingtin gene. The peripheral innate immune system is dysregulated in Huntington’s disease and may contribute to its pathogenesis. However, it is not clear whether or to what extent the adaptive immune system is also involved. Here, we carry out the first comprehensive investigation of human ex vivo T lymphocytes in Huntington’s disease, focusing on the frequency of a range of T lymphocyte subsets, as well as analysis of proliferation, cytokine production and gene transcription. In contrast to the innate immune system, the intrinsic phenotype of T lymphocytes does not appear to be affected by the presence of mutant huntingtin, with Huntington’s disease T lymphocytes exhibiting no significant functional differences compared to control cells. The transcriptional profile of T lymphocytes also does not appear to be significantly affected, suggesting that peripheral immune dysfunction in Huntington’s disease is likely to be mediated primarily by the innate rather than the adaptive immune system. This study increases our understanding of the effects of Huntington’s disease on peripheral tissues, while further demonstrating the differential effects of the mutant protein on different but related cell types. Finally, this study suggests that the potential use of novel therapeutics aimed at modulating the Huntington’s disease innate immune system should not be extended to include the adaptive immune system.
Collapse
Affiliation(s)
- James R. C. Miller
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Ulrike Träger
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Ralph Andre
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Sarah J. Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Ramsingh AI, Manley K, Rong Y, Reilly A, Messer A. Transcriptional dysregulation of inflammatory/immune pathways after active vaccination against Huntington's disease. Hum Mol Genet 2015; 24:6186-97. [PMID: 26307082 PMCID: PMC4599676 DOI: 10.1093/hmg/ddv335] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy, both active and passive, is increasingly recognized as a powerful approach to a wide range of diseases, including Alzheimer's and Parkinson's. Huntington's disease (HD), an autosomal dominant disorder triggered by misfolding of huntingtin (HTT) protein with an expanded polyglutamine tract, could also benefit from this approach. Individuals can be identified genetically at the earliest stages of disease, and there may be particular benefits to a therapy that can target peripheral tissues in addition to brain. In this active vaccination study, we first examined safety and immunogenicity for a broad series of peptide, protein and DNA plasmid immunization protocols, using fragment (R6/1), and knock-in (zQ175) models. No safety issues were found. The strongest and most uniform immune response was to a combination of three non-overlapping HTT Exon1 coded peptides, conjugated to KLH, delivered with alum adjuvant. An N586-82Q plasmid, delivered via gene gun, also showed ELISA responses, mainly in the zQ175 strain, but with more variability, and less robust responses in HD compared with wild-type controls. Transcriptome profiling of spleens from the triple peptide-immunized cohort showed substantial HD-specific differences including differential activation of genes associated with innate immune responses, absence of negative feedback control of gene expression by regulators, a temporal dysregulation of innate immune responses and transcriptional repression of genes associated with memory T cell responses. These studies highlight critical issues for immunotherapy and HD disease management in general.
Collapse
Affiliation(s)
- Arlene I Ramsingh
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Kevin Manley
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Yinghui Rong
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Andrew Reilly
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and
| | - Anne Messer
- New York State Department of Health, Wadsworth Center, Albany, NY, USA and Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY, USA
| |
Collapse
|
50
|
The Potential Regulatory Mechanisms of miR-196a in Huntington's Disease through Bioinformatic Analyses. PLoS One 2015; 10:e0137637. [PMID: 26376480 PMCID: PMC4574104 DOI: 10.1371/journal.pone.0137637] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/20/2015] [Indexed: 11/19/2022] Open
Abstract
High throughput screening is a powerful tool to identify the potential candidate molecules involved during disease progression. However, analysis of complicated data is one of the most challenging steps on the way to obtaining useful results from this approach. Previously, we showed that a specific miRNA, miR-196a, could ameliorate the pathological phenotypes of Huntington’s disease (HD) in different models, and performed high throughput screening by using the striatum of transgenic mice. In this study, we further tried to identify the potential regulatory mechanisms using different bioinformatic tools, including Database for Annotation, Visualization and Integrated Discovery (DAVID), Molecular Signatures Database (MSigDB), TargetScan and MetaCore. The results showed that miR-196a dominantly altered “ABC transporters”, “RIG-I-like receptor signaling pathway”, immune system”, “adaptive immune system”,“tissue remodeling and wound repair” and “cytoskeleton remodeling”. In addition, miR-196a also changed the expression of several well-defined pathways of HD, such as apoptosis and cell adhesion. Since these analyses showed the regulatory pathways are highly related to the modification of the cytoskeleton, we further confirmed that miR-196a could enhance the neurite outgrowth in neuroblastoma cells, suggesting miR-196a might provide beneficial functions through the alteration of cytoskeleton structures. Since impairment of the cytoskeleton has been reported in several neuronal diseases, this study will provide not only the potential working mechanisms of miR-196a but also insights for therapeutic strategies for use with different neuronal diseases.
Collapse
|