1
|
Xing C, Chen H, Bi W, Lei T, Hang Z, Du H. Targeting 5-HT Is a Potential Therapeutic Strategy for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:13446. [PMID: 39769209 PMCID: PMC11679250 DOI: 10.3390/ijms252413446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
There is increasing interest in the potential therapeutic role of 5-HT (serotonin) in the treatment of neurodegenerative diseases, which are characterized by the progressive degeneration and death of nerve cells. 5-HT is a vital neurotransmitter that plays a central role in regulating mood, cognition, and various physiological processes in the body. Disruptions in the 5-HT system have been linked to several neurological and psychiatric disorders, making it an attractive target for therapeutic intervention. Although the exact causes of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are not fully understood, researchers believe that regulating the 5-HT system could help alleviate symptoms and potentially slow the progression of these diseases. Here, we delve into the potential of harnessing 5-HT as a therapeutic target for the treatment of neurodegenerative diseases. It is important to note that the current clinical drugs targeting 5-HT are still limited in the treatment of these complex diseases. Therefore, further research and clinical trials are needed to evaluate the feasibility and effectiveness of its clinical application.
Collapse
Affiliation(s)
- Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongyu Chen
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Wangyu Bi
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Zhongci Hang
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongwu Du
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| |
Collapse
|
2
|
Jellinger KA. The pathobiology of depression in Huntington's disease: an unresolved puzzle. J Neural Transm (Vienna) 2024; 131:1511-1522. [PMID: 38349403 DOI: 10.1007/s00702-024-02750-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 12/01/2024]
Abstract
Huntington's disease (HD) is an autosomal-dominant progressive neurodegenerative disease that manifests with a triad of symptoms including motor dysfunctions, cognitive deficits, and prominent neuropsychiatric symptoms, the most common of which is depression, with a prevalence between 30 and 70%. Depressive symptoms occur in all stages of HD, beginning in presymptomatic HD gene carriers, and are strongly associated with suicidal ideation and suicidality, but their relationship with other clinical dimensions in HD is controversial and the underlying pathophysiology is poorly understood. Analysis of the available literature until November 2023 concerned the prevalence, clinical manifestations, neuroimaging, transgenic models, and treatment options of HD depression. While it was believed that depression in HD is due to psychosomatic factors in view of the fatal disease, studies in transgenic models of HD demonstrated molecular changes including neurotrophic and serotonergic dysregulation and disorders of the hypothalamic-pituitary-adrenal axis inducing depression-like changes. While relevant neuropathological data are missing, recent neuroimaging studies revealed correlations between depressive symptoms and dysfunctional connectivities in the default mode network, basal ganglia and prefrontal cortex, and changes in limbic and paralimbic structures related to the basic neurodegenerative process. The impact of response to antidepressants in HD patients is controversial; selective serotonin reuptake inhibitors are superior to serotonin-norepinephrine reuptake inhibitors, while electroconvulsive therapy may be effective for pharmacotherapy resistant cases. Since compared to major depressive disorder and depression in other neurodegenerative diseases, our knowledge of the molecular basis in HD depression is limited, further studies to elucidate the heterogeneous pathogenesis in this fatal disorder are warranted.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
3
|
Mora I, Teixidó A, Vázquez-Manrique RP, Puiggròs F, Arola L. Docosahexaenoic Acid (DHA) Supplementation in a Triglyceride Form Prevents from Polyglutamine-Induced Dysfunctions in Caenorhabditis elegans. Int J Mol Sci 2024; 25:12594. [PMID: 39684306 DOI: 10.3390/ijms252312594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
A common hallmark of neurodegenerative diseases is the accumulation of polypeptide aggregates in neurons. Despite the primary cause of these diseases being inherently genetic, their development can be delayed with proper preventive treatments. Long-chain polyunsaturated fatty acids (ω-3 LCPUFA) are promising bioactive nutrients that are beneficial for brain health. In this study, the impact of an oil rich in a structured form of docosahexaenoic acid (DHA) triglyceride (TG) was assessed in a Caenorhabditis elegans model expressing long poly-glutamine (polyQ) chains, which mimics the symptomatology of polyQ-related neurodegenerative diseases such as Huntington's disease (HD), among others. The lifespan, the motility, the number of polyQ aggregates, the oxidative stress resistance, and the cognitive performance associated with sensitive stimuli was measured in mutant nematodes with polyQ aggregates. Overall, DHA-TG at 0.5 µM improved the lifespan, the motility, the oxidative stress resistance, and the cognitive performance of the nematodes, emphasizing the protection against serotonergic synapse dysfunction. Furthermore, the treatment reduced the polyQ aggregates in the nematodes. The data described herein shed light on the connection between DHA and the cognitive performance in neurodegenerative diseases and demonstrated the potential of DHA-TG as nutritional co-adjuvant to prevent the development of polyQ-associated dysfunctions.
Collapse
Affiliation(s)
- Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain
- Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Alex Teixidó
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health Unit, 43204 Reus, Spain
| | - Rafael P Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Joint Unit for Rare Diseases, Insituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Tarragona, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| |
Collapse
|
4
|
van Wamelen DJ, Martin NH, Makos O, Badenoch J, Valera-Bermejo JM, Hartmann M, Cristales AR, Wood TC, Veronese M, Moretto M, Zelaya F, dell'Acqua F, O'Daly O, Lythgoe DJ, Ginestet C, Turkheimer F, Palasits N, Mrzljak L, Warner JH, Rabiner EA, Gunn R, Tabrizi SJ, Sampaio C, Wood A, Williams SC. Study protocol for the iMarkHD study in individuals with Huntington's disease. J Huntingtons Dis 2024; 13:479-489. [PMID: 39973385 DOI: 10.1177/18796397241288165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background: Huntington's disease (HD) is still often defined by the onset of motor symptoms, inversely associated with the size of the CAG repeat expansion in the huntingtin gene. Although the cause of HD is known, much remains unknown about mechanisms underlying clinical symptom development, disease progression, and specific clinical subtypes/endophenotypes. Objective: In the iMarkHD study, we aim to investigate four discrete molecular positron emission tomography (PET) tracers and magnetic resonance imaging (MRI) markers as biomarkers for disease and symptom progression. Methods: Following MRI optimization in five healthy volunteers (cohort 1), we aim to recruit 108 participants of whom 72 are people with HD (PwHD) and 36 healthy volunteers (cohort 2). Pending interim analysis, these numbers could increase to 96 PwHD and 48 healthy controls. Participants will complete a total of 10 study visits, consisting of a screening visit followed by a clinical and MRI visit and PET visits at baseline, year 1, and year 2. PET targets include the cannabinoid 1, histamine 3, and serotonin 2A receptors, and phosphodiesterase 10A, whereas MRI will be multimodal, including, but not limited to, the assessment of cerebral blood flow, functional connectivity, and brain iron. Results: Recruitment is currently active and started in September 2022. Conclusions: By combining PET and multi-modal MRI assessments we expect to provide a comprehensive examination of the molecular, functional, and structural framework of HD progression. As such, the iMarkHD study will provide a solid base for the identification of treatment targets and novel outcome measures for future clinical trials.
Collapse
Affiliation(s)
- Daniel J van Wamelen
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, the Netherlands
| | - Naomi H Martin
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Orsolya Makos
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - James Badenoch
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Jose Manuel Valera-Bermejo
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Monika Hartmann
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Alay Rangel Cristales
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Tobias C Wood
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Mattia Veronese
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Manuela Moretto
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Fernando Zelaya
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Flavio dell'Acqua
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Owen O'Daly
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - David J Lythgoe
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Cedric Ginestet
- Institute of Psychiatry, Psychology & Neuroscience, Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Federico Turkheimer
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Nikki Palasits
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Ladislav Mrzljak
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | - John H Warner
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | | | - Roger Gunn
- Invicro, A Konica Minolta Company, London, UK
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, UK Dementia Research Institute, Department of Neurodegenerative Diseases, University College London, London, UK
| | - Cristina Sampaio
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Andrew Wood
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Steven Cr Williams
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| |
Collapse
|
5
|
Marinina KS, Bezprozvanny IB, Egorova PA. Anxiety, memory, and social impairments in the YAC128 mouse model of Huntington's disease. J Huntingtons Dis 2024; 13:431-448. [PMID: 39973387 DOI: 10.1177/18796397241295668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Huntington's disease (HD) is an autosomal dominant hereditary disorder, caused by an expansion of polyglutamine in the huntingtin protein. HD is characterized by a progressive decline in motor functions. This decline includes involuntary movements (chorea) and the worsening of controlled motions caused mainly by neuronal dysfunction in the striatum. In addition to the deterioration of motor symptoms, HD patients also suffer from cognitive changes, mood swings, apathy, depression, outbursts of anger, psychosis, and social withdrawal. OBJECTIVE A comprehensive examination of cognitive, affective, and social changes in the HD mouse model is required for the development of combined therapy for both motor and non-motor deficits in HD. METHODS The behavioral tests for anxiety, memory, and social traits were used in this study. RESULTS YAC128 HD transgenic mice exhibited anxiolytic behavior in the novel brightly illuminated environment of the open field and light-dark place preference tests. Moreover, YAC128 HD mice also suffered from a decline in their recognition memory during the novel object recognition test. YAC128 HD mice demonstrated reduced exploration interest during the open field with a non-social target as well as during the first day of the three-chamber social test. Social interaction was also impaired in YAC128 HD mice as it was shown in the social interaction with resident intruder test. CONCLUSIONS YAC128 HD mouse model may be used as a model system to test the possible treatments for both motor and non-motor symptoms including memory loss, agitation and social withdrawal.
Collapse
Affiliation(s)
- Ksenia S Marinina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Laboratory of Mechanisms of Regulation and Compensation of the Nervous System Pathological Excitability, Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, St. Petersburg, Russia
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Polina A Egorova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
6
|
Zadegan SA, Ramirez F, Reddy KS, Sahin O, Rocha NP, Teixeira AL, Furr Stimming E. Treatment of Depression in Huntington's Disease: A Systematic Review. J Neuropsychiatry Clin Neurosci 2024; 36:283-299. [PMID: 38528808 DOI: 10.1176/appi.neuropsych.20230120] [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] [Indexed: 03/27/2024]
Abstract
Depression is a common psychiatric disorder among individuals with Huntington's disease (HD). Depression in HD and major depressive disorder appear to have different pathophysiological mechanisms. Despite the unique pathophysiology, the treatment of depression in HD is based on data from the treatment of major depressive disorder in the general population. The objective of this systematic review was to conduct a comprehensive evaluation of the available evidence. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Studies on the treatment of depression in HD were identified by searching MEDLINE, Embase, and PsycInfo. The initial search yielded 2,771 records, 41 of which were ultimately included. There were 19 case reports, seven case series, three cross-sectional studies, one qualitative study, nine nonrandomized studies, and two randomized trials among the included studies. The most common assessment tools were the Hospital Anxiety and Depression Scale (N=8), the Beck Depression Inventory (N=6), and the Hamilton Depression Rating Scale (N=6). Only 59% of the included studies assessed depressive symptoms with a scoring system. The pharmacological options for the treatment of depression included antidepressants and antipsychotics. Nonpharmacological approaches were multidisciplinary rehabilitation, psychotherapy, and neurostimulation. Limited evidence on the treatment of depression in HD was available, and this literature consisted mainly of case reports and case series. This systematic review highlights the knowledge gap and the pressing need for HD-specific research to determine the efficacy of treatment approaches for depression in HD.
Collapse
Affiliation(s)
- Shayan Abdollah Zadegan
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Frank Ramirez
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Kirthan S Reddy
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Onur Sahin
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Natalia Pessoa Rocha
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Antonio L Teixeira
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| | - Erin Furr Stimming
- Department of Neurology (Zadegan, Ramirez, Rocha, Furr Stimming) and Department of Psychiatry and Behavioral Sciences (Teixeira), McGovern Medical School (Reddy, Sahin), University of Texas Health Science Center at Houston; Huntington's Disease Society of America Center of Excellence at University of Texas Health Science Center at Houston (Zadegan, Ramirez, Rocha, Teixeira, Furr Stimming)
| |
Collapse
|
7
|
Gubert C, Kong G, Costello C, Adams CD, Masson BA, Qin W, Choo J, Narayana VK, Rogers G, Renoir T, Furness JB, Hannan AJ. Dietary fibre confers therapeutic effects in a preclinical model of Huntington's disease. Brain Behav Immun 2024; 116:404-418. [PMID: 38142919 DOI: 10.1016/j.bbi.2023.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder involving psychiatric, cognitive and motor deficits, as well as peripheral symptoms, including gastrointestinal dysfunction. The R6/1 HD mouse model expresses a mutant human huntingtin transgene and has been shown to provide an accurate disease model. Recent evidence of gut microbiome disruption was shown in preclinical and clinical HD. Therefore, we aimed to assess the potential role of gut microbial modulation in the treatment of HD. The R6/1 HD mice and wild-type littermate controls were randomised to receive diets containing different amounts of fibre: high-fibre (10 % fibre), control (5 % fibre), or zero-fibre (0 % fibre), from 6 to 20 weeks of age. We characterized the onset and progression of motor, cognitive and affective deficits, as well as gastrointestinal function and gut morphological changes. Faeces were collected for gut microbiome profiling using 16S rRNA sequencing, at 14 and 20 weeks of age. When compared to the control diet, high-fibre diet improved the performance of HD mice in behavioral tests of cognitive and affective function, as well as the gastrointestinal function of both HD and wild-type mice. While the diets changed the beta diversity of wild-type mice, no statistical significance was observed at 14 or 20 weeks of age within the HD mice. Analysis of Composition of Microbiomes with Bias Correction (ANCOM-BC) models were performed to evaluate microbiota composition, which identified differences, including a decreased relative abundance of the phyla Actinobacteriota, Campylobacterota and Proteobacteria and an increased relative abundance of the families Bacteroidaceae, Oscillospiraceae and Ruminococcaceae in HD mice when compared to wild-type mice after receiving high-fibre diet. PICRUSt2 revealed that high-fibre diet also decreased potentially pathogenic functional pathways in HD. In conclusion, high-fibre intake was effective in enhancing gastrointestinal function, cognition and affective behaviors in HD mice. These findings indicate that dietary fibre interventions may have therapeutic potential in Huntington's disease to delay clinical onset, and have implications for related disorders exhibiting dysfunction of the gut-brain axis.
Collapse
Affiliation(s)
- Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Geraldine Kong
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Peter Doherty Institute of Infection and Immunity, University of Melbourne, Parkville, Victoria 3000, Australia
| | - Callum Costello
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Cameron D Adams
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jocelyn Choo
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Vinod K Narayana
- Metabolomics Australia Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Geraint Rogers
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville 3010, Australia
| | - John B Furness
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville 3010, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
8
|
Bilal H, Harding IH, Stout JC. The relationship between disease-specific psychosocial stressors and depressive symptoms in Huntington's disease. J Neurol 2024; 271:289-299. [PMID: 37695532 PMCID: PMC10769991 DOI: 10.1007/s00415-023-11982-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Huntington's disease (HD) is an inherited neurodegenerative disease involving motor abnormalities, cognitive decline, and psychological difficulties. Depression is among the most common psychological difficulties in HD. People with HD encounter numerous stressors related to their diagnosis and the impact of HD on their daily lives. Understanding the relationship between HD-specific psychosocial stressors and depression symptoms is critical for optimising treatment and developing a holistic, disease-specific model of depression in HD. METHODS Fifty-seven adults with the HD gene expansion (33 pre-symptomatic, 24 symptomatic) completed a self-report depression questionnaire and rated how much stress they experienced in relation to 20 psychosocial challenges commonly associated with HD. We examined associations between depression symptoms and each stressor individually, and after clustering using principal components analysis. RESULTS Depression symptoms were significantly associated with most of the psychosocial stressors assessed. Clustering with principal components analysis revealed that higher depression scores had significant independent associations with greater stress related to the future implications of HD (β = .44, p = .001) and sleep and psychological difficulties (β = .28, p = .005), but not with stress related to functional limitations (β = .11, p = .33) or interpersonal issues caused by HD (β = .15, p = .21). CONCLUSIONS Stressful experiences associated with HD constitute an important risk factor for depression in HD. Our findings support the use of more psychologically informed models of depression in HD and necessitate further research on tailored psychosocial interventions for HD patients with depression.
Collapse
Affiliation(s)
- Hiba Bilal
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, 18 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Ian H Harding
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, 18 Innovation Walk, Clayton, VIC, 3800, Australia.
| |
Collapse
|
9
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Ferrari C, Capacci E, Bagnoli S, Ingannato A, Sorbi S, Nacmias B. The Huntington's Disease Gene in an Italian Cohort of Patients with Bipolar Disorder. Genes (Basel) 2023; 14:1681. [PMID: 37761821 PMCID: PMC10531383 DOI: 10.3390/genes14091681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Huntington's disease (HD) is characterized by motor, cognitive and psychiatric manifestations and caused by an expansion of CAG repeats over 35 triplets on the huntingtin (HTT) gene. However, expansions in the range 27-35 repeats (intermediate allele) can be associated with pathological phenotypes. The onset of HD is conventionally defined by the onset of motor symptoms, but psychiatric disturbances can precede the motor phase by up to twenty years. The aims of the present study are to identify HD patients in the pre-motor phase of the disease among patients diagnosed with bipolar disorders and evaluate any differences between bipolar patients carrying the normal HTT allele and patients with the expanded HTT gene. METHODS We assessed the HTT genotype in an Italian cohort of 69 patients who were affected by either type 1 or type 2 bipolar disorder. RESULTS No patient was found to be a carrier of the pathological HTT allele, but 10% of bipolar subjects carried an intermediate allele. Carriers of the intermediate allele were older at the onset of psychiatric symptoms than non-carriers. CONCLUSION The pathological HTT gene was not associated with bipolar disorder, while we found a higher frequency of the intermediate allele among the bipolar population with respect to healthy controls. The identification of this subset of bipolar subjects has implications for the clinical management of patients and their family members and promotes further investigation into possible pathological mechanisms common to both HD and bipolar disorder.
Collapse
Affiliation(s)
- Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
| | - Elena Capacci
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, 50143 Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy; (E.C.); (S.B.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, 50143 Florence, Italy
| |
Collapse
|
11
|
Zalewska T, Pawelec P, Ziabska K, Ziemka-Nalecz M. Sexual Dimorphism in Neurodegenerative Diseases and in Brain Ischemia. Biomolecules 2022; 13:26. [PMID: 36671411 PMCID: PMC9855831 DOI: 10.3390/biom13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in brain responses to several neurological conditions. It is suggested that sex-related differences between men and women may have profound effects on disease susceptibility, pathophysiology, and progression. Sexual differences of the brain are achieved through the complex interplay of several factors contributing to this phenomenon, such as sex hormones, as well as genetic and epigenetic differences. Despite recent advances, the precise link between these factors and brain disorders is incompletely understood. This review aims to briefly outline the most relevant aspects that differ between men and women in ischemia and neurodegenerative disorders (AD, PD, HD, ALS, and SM). Recognition of disparities between both sexes could aid the development of individual approaches to ameliorate or slow the progression of intractable disorders.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Str., 02-106 Warsaw, Poland
| | | | | | | |
Collapse
|
12
|
Xu C, Su L, Qiu N, Hou M, Yu F, Zou X, Wang J. The Effect of Unpredictable Chronic Stress on Rare Minnow ( Gobiocypris rarus): Growth, Behaviour and Physiology. BIOLOGY 2022; 11:1755. [PMID: 36552265 PMCID: PMC9775413 DOI: 10.3390/biology11121755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
Fishes often adjust their behaviour patterns and physiological responses to cope with changing environments, and different life experiences affect them differently. Fishes might adapt to short-term stress, whereas long-term unpredictable stress may lead to various adverse effects. Although some studies have constructed unpredictable stress models of fish, the effect of unpredictable chronic stress (UCS) in the laboratory is poorly understood in fishes. In the current study, we exposed adult rare minnow to an unpredictable chronic stress protocol over 7 and 14 days and measured their response in terms of growth performance, cortisol, neurotransmitter levels (DA, 5-HT, and related metabolites), and behaviour patterns to comprehensively assess the effects of UCS on laboratory rare minnow. We discovered that specific growth rates were significantly decreased, and cortisol levels were lowered in both 7-days and 14-days stress groups. In the behaviour test, the activity level of the 14-days stress group increased, but there was no significant difference in the number of crossings to the center areas, time spent in the center areas, or the speed. In addition, the levels of DA and 5-HT did not change in the stress groups, but the DOPAC and 5-HIAA levels in the 14 days stress group were significantly higher than those in the control group. These results suggested that UCS influences rare minnow growth performance, behaviour patterns, and cortisol levels, and similar stress should be minimised in the laboratory.
Collapse
Affiliation(s)
- Chunsen Xu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liangxia Su
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Ning Qiu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Miaomiao Hou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fandong Yu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinhua Zou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianwei Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- National Aquatic Biological Resource Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430070, China
| |
Collapse
|
13
|
Mees I, Li S, Tran H, Ang CS, Williamson NA, Hannan AJ, Renoir T. Phosphoproteomic dysregulation in Huntington's disease mice is rescued by environmental enrichment. Brain Commun 2022; 4:fcac305. [PMID: 36523271 PMCID: PMC9746689 DOI: 10.1093/braincomms/fcac305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 09/05/2022] [Accepted: 11/21/2022] [Indexed: 09/05/2023] Open
Abstract
Huntington's disease is a fatal autosomal-dominant neurodegenerative disorder, characterized by neuronal cell dysfunction and loss, primarily in the striatum, cortex and hippocampus, causing motor, cognitive and psychiatric impairments. Unfortunately, no treatments are yet available to modify the progression of the disease. Recent evidence from Huntington's disease mouse models suggests that protein phosphorylation (catalysed by kinases and hydrolysed by phosphatases) might be dysregulated, making this major post-translational modification a potential area of interest to find novel therapeutic targets. Furthermore, environmental enrichment, used to model an active lifestyle in preclinical models, has been shown to alleviate Huntington's disease-related motor and cognitive symptoms. However, the molecular mechanisms leading to these therapeutic effects are still largely unknown. In this study, we applied a phosphoproteomics approach combined with proteomic analyses on brain samples from pre-motor symptomatic R6/1 Huntington's disease male mice and their wild-type littermates, after being housed either in environmental enrichment conditions, or in standard housing conditions from 4 to 8 weeks of age (n = 6 per group). We hypothesized that protein phosphorylation dysregulations occur prior to motor onset in this mouse model, in two highly affected brain regions, the striatum and hippocampus. Furthermore, we hypothesized that these phosphoproteome alterations are rescued by environmental enrichment. When comparing 8-week-old Huntington's disease mice and wild-type mice in standard housing conditions, our analysis revealed 229 differentially phosphorylated peptides in the striatum, compared with only 15 differentially phosphorylated peptides in the hippocampus (statistical thresholds fold discovery rate 0.05, fold change 1.5). At the same disease stage, minor differences were found in protein levels, with 24 and 22 proteins dysregulated in the striatum and hippocampus, respectively. Notably, we found no differences in striatal protein phosphorylation and protein expression when comparing Huntington's disease mice and their wild-type littermates in environmentally enriched conditions. In the hippocampus, only four peptides were differentially phosphorylated between the two genotypes under environmentally enriched conditions, and 22 proteins were differentially expressed. Together, our data indicates that protein phosphorylation dysregulations occur in the striatum of Huntington's disease mice, prior to motor symptoms, and that the kinases and phosphatases leading to these changes in protein phosphorylation might be viable drug targets to consider for this disorder. Furthermore, we show that an early environmental intervention was able to rescue the changes observed in protein expression and phosphorylation in the striatum of Huntington's disease mice and might underlie the beneficial effects of environmental enrichment, thus identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Isaline Mees
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
| | - Harvey Tran
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
14
|
Hannan AJ. Chorea me a river: depression in Huntington's disease as an exemplar of precision medicine. Brain Commun 2022; 4:fcac294. [PMID: 36440099 PMCID: PMC9683389 DOI: 10.1093/braincomms/fcac294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/14/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
This scientific commentary refers to 'Different depression: motivational anhedonia governs antidepressant efficacy in Huntington's disease' by McLauchlan et al. (https://doi.org/10.1093/braincomms/fcac278).
Collapse
Affiliation(s)
- Anthony J Hannan
- Correspondence to: Prof. Anthony J. Hannan, Florey Institute of
Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia.
E-mail:
| |
Collapse
|
15
|
Weng YT, Chen HM, Chien T, Chiu FL, Kuo HC, Chern Y. TRAX Provides Neuroprotection for Huntington's Disease Via Modulating a Novel Subset of MicroRNAs. Mov Disord 2022; 37:2008-2020. [PMID: 35997316 DOI: 10.1002/mds.29174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/19/2022] [Accepted: 07/14/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is a neurodegenerative disease caused by CAG-repeat expansions (>36) in exon 1 of HTT, which dysregulates multiple cellular machineries. Translin-associated protein X (TRAX) is a scaffold protein with diverse functions, including suppressing the microRNA (miRNA)-mediated silencing by degrading pre-miRNA. To date, the role of TRAX in neurodegenerative diseases remains unknown. OBJECTIVES We delineated the role of TRAX upregulation during HD progression. METHODS Expression of TRAX in the brains of humans and three mouse models with HD were analyzed by immunohistochemistry staining, western blot, and quantitative reverse transcription-polymerase chain reaction. Adeno-associated viruses harboring TRAX short hairpin RNA were intrastriatally injected into HD mice to downregulate TRAX. HD-like symptoms were analyzed by behavioral and biochemical assessments. The miRNA-sequencing and RNA-sequencing analyses were used to identify the TRAX- regulated miRNA-messenger RNA (mRNA) axis during HD progression. The identified gene targets were validated biochemically in mouse and human striatal cells. RESULTS We discovered that TRAX was upregulated in the brains of HD patients and three HD mouse models. Downregulation of TRAX enhanced 83 miRNAs (including miR-330-3p, miR-496a-3p) and subsequently changed the corresponding mRNA networks critical for HD pathogenesis (eg, DARPP-32 and brain-derived neurotrophic factor). Disruption of the TRAX-mediated miRNA-mRNA axis accelerated the progression of HD-like symptoms, including the degeneration of motor function, accumulation of mHTT aggregates, and shortened neurite outgrowth. CONCLUSIONS We demonstrated that TRAX upregulation is authentic and protective in HD. Our study provides a novel layer of regulation for HD pathogenesis and may lead to the development of new therapeutic strategies for HD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yu-Ting Weng
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ting Chien
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Feng-Lan Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
16
|
Brait VH, Jackman KA, Pang TY. Effects of wheel-running on anxiety and depression-relevant behaviours in the MCAO mouse model of stroke: moderation of brain-derived neurotrophic factor and serotonin receptor gene expression. Behav Brain Res 2022; 432:113983. [PMID: 35777551 DOI: 10.1016/j.bbr.2022.113983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/06/2022] [Accepted: 06/27/2022] [Indexed: 11/02/2022]
Abstract
Stroke continues to be a major cause of mortality globally. Post-stroke treatment is complicated by the heterogenous nature of pathology and the emergence of secondary psychological symptoms are an additional challenge to the recovery process. Poststroke depression (PSD) is a common co-morbidity and is a major impediment to recovery. While selective serotonin reuptake inhibitors (SSRIs) have proven to be clinically efficacious in treating PSD, the pathogenic processes that underlie the manifestation of depressive mood post-stroke remains unclear. Furthermore, the use of SSRIs is associated with risks of intracerebral haemorrhage, so alternative treatment options need to be continuously explored. Exercise has been demonstrated to be beneficial for improving mood in humans and preclinical models of neurological conditions. Little is known of the mood-related benefits of physical exercise post-stroke. Using the middle cerebral artery occlusion (MCAO) mouse model of cerebral ischaemia, we investigated whether behavioural deficits emerge post-MCAO and could be rescued by voluntary wheel-running. We report that MCAO induced hypo-locomotion and anhedonia-related behaviours, with some improvements conferred by wheel-running. Serotonin transporter gene expression was increased in the MCAO hippocampus and frontal cortex, but this increase remained despite wheel-running. Wheel-running associated up-regulation of BDNF gene expression was unaffected in MCAO mice, reflecting conservation of key neuroplasticity molecular pathways. Taken together, our results highlight the need for further research into serotonergic modulation of the affective symptoms of stroke.
Collapse
Affiliation(s)
- Vanessa H Brait
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Katherine A Jackman
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Terence Y Pang
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia; Department of Anatomy and Physiology, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
17
|
Environmental stimulation in Huntington disease patients and animal models. Neurobiol Dis 2022; 171:105725. [DOI: 10.1016/j.nbd.2022.105725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 01/07/2023] Open
|
18
|
The Effect of Environmental Enrichment on Laboratory Rare Minnows (Gobiocypris rarus): Growth, Physiology, and Behavior. Animals (Basel) 2022; 12:ani12040514. [PMID: 35203222 PMCID: PMC8868387 DOI: 10.3390/ani12040514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Environmental enrichment is an important part of animal welfare. In this study, rare minnow in different rearing conditions underwent comprehensive evaluation regarding growth, anxiety-like behavior, and physiology parameters. Results showed that there were no differences in SGR, anxiety-like behavior, DA, DOPAC, and 5-HIAA levels between control and enriched groups. However, the enriched group had higher cortisol and 5-HT levels. Therefore, researchers should focus on the effect of environmental enrichment regarding the welfare of rare minnow and how it effects the validity of data from laboratory studies. Abstract Environmental enrichment is a method to increase environmental heterogeneity, which may reduce stress and improve animal welfare. Previous studies have shown that environmental enrichment can increase the growth rate, decrease aggressive and anxiety-like behaviors, improve learning ability and agility, and reduce cortisol levels in animals. These effects usually differ between species. Unfortunately, habitat enrichment on laboratory fish is poorly studied and seldom adopted in care guidance. Rare minnows (Gobiocypris rarus) have been cultured as a native laboratory fish in China in barren banks without environmental enrichment since 1990; they have been widely used in studies on ecotoxicology, environmental science, and other topics. The purpose of this study was to investigate the effect of environment enrichment on the growth, physiological status, and anxiety-like behavior of laboratory rare minnows. We observed and analyzed SGR, cortisol levels, DA, DOPAC, 5-HT and 5-HIAA, and anxiety-like behavior indexes after one month of treatment in barren (control) and enrichment tanks. We found that there were no significant differences in SGR, anxiety-like behavior, DA, DOPAC, or 5-HIAA levels between the two treatments. However, higher cortisol and 5-HT levels were observed in the enrichment tanks. This study suggests that rare minnows might be influenced by their living environment, and future related studies should consider their environmental enrichment.
Collapse
|
19
|
McLauchlan DJ, Lancaster T, Craufurd D, Linden DEJ, Rosser AE. Different depression: motivational anhedonia governs antidepressant efficacy in Huntington's disease. Brain Commun 2022; 4:fcac278. [PMID: 36440100 PMCID: PMC9683390 DOI: 10.1093/braincomms/fcac278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/13/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Depression is more common in neurodegenerative diseases such as Huntington's disease than the general population. Antidepressant efficacy is well-established for depression within the general population: a recent meta-analysis showed serotonin norepinephrine reuptake inhibitors, tricyclic antidepressants and mirtazapine outperformed other antidepressants. Despite the severe morbidity, antidepressant choice in Huntington's disease is based on Class IV evidence. We used complementary approaches to determine treatment choice for depression in Huntington's disease: propensity score analyses of antidepressant treatment outcome using the ENROLL-HD data set, and a dissection of the cognitive mechanisms underlying depression in Huntington's disease using a cognitive battery based on the Research Domain Criteria for Depression. Study 1 included ENROLL-HD 5486 gene-positive adult patients started on an antidepressant medication for depression. Our outcome measures were depression (Hospital Anxiety and Depression Scale or Problem Behaviours Assessment 'Depressed Mood' item) at first follow-up (primary outcome) and all follow-ups (secondary outcome). The intervention was antidepressant class. We used Svyglm&Twang in R to perform propensity scoring, using known variables (disease progression, medical comorbidity, psychiatric morbidity, sedatives, number of antidepressants, demographics and antidepressant contraindications) to determine the probability of receiving different antidepressants (propensity score) and then included the propensity score in a model of treatment efficacy. Study 2 recruited 51 gene-positive adult patients and 26 controls from the South Wales Huntington's Disease Management Service. Participants completed a motor assessment, in addition to measures of depression and apathy, followed by tasks measuring consummatory anhedonia, motivational anhedonia, learning from reward and punishment and reaction to negative outcome. We used generalised linear models to determine the association between task performance and depression scores. Study 1 showed selective serotonin reuptake inhibitors outperformed serotonin norepinephrine reuptake inhibitors on the primary outcome (P = 0.048), whilst both selective serotonin reuptake inhibitors (P = 0.00069) and bupropion (P = 0.0045) were superior to serotonin norepinephrine reuptake inhibitors on the secondary outcome. Study 2 demonstrated an association between depression score and effort for reward that was not explained by apathy. No other mechanisms were associated with depression score. We found that selective serotonin reuptake inhibitors and bupropion outperform serotonin norepinephrine reuptake inhibitors at alleviating depression in Huntington's disease. Moreover, motivational anhedonia appears the most significant mechanism underlying depression in Huntington's disease. Bupropion is improves motivational anhedonia and has a synergistic effect with selective serotonin reuptake inhibitors. This work provides the first large-scale, objective evidence to determine treatment choice for depression in Huntington's disease, and provides a model for determining antidepressant efficacy in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Duncan James McLauchlan
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Neurology, Morriston Hospital, Swansea Bay University Health Board, Swansea SA6 6NL, UK
| | - Thomas Lancaster
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Psychology, University of Bath, Bath BA2 7AY, UK
| | - David Craufurd
- Manchester Center for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Center, Manchester M13 9PL, UK.,St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Center, Manchester M13 9WL, UK
| | - David E J Linden
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Psychology, University of Bath, Bath BA2 7AY, UK.,School for Mental Health and Neuroscience, Fac. Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Anne E Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Neurology, Morriston Hospital, Swansea Bay University Health Board, Swansea SA6 6NL, UK.,School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
20
|
The underestimated sex: a review on female animal models of depression. Neurosci Biobehav Rev 2021; 133:104498. [PMID: 34953920 DOI: 10.1016/j.neubiorev.2021.12.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023]
Abstract
Major depression (MD) is the most common psychiatric disorder, predicted to affect around 264 million people worldwide. Although the etiology of depression remains elusive, the interplay between genetics and environmental factors, such as early life events, stress, exposure to drugs and health problems appears to underlie its development. Whereas depression is twice more prevalent in women than in men, most preclinical studies are performed in male rodents. In fact, females' physiology and reproductive experience are associated with changes to brain, behavior and endocrine profiles that may influence both stress, an important precipitating factor for depression, and response to treatment. These specificities emphasize the need to choose the most suitable models and readouts in order to better understand the pathophysiological mechanisms of depression in females. With this review, we aim to provide an overview of female animal models of depression highlighting the major differences between models, regarding behavioral, physiological, and molecular readouts, but also the major gaps in research, attending to the role of etiological factors, protocol variability and sex.
Collapse
|
21
|
Akimov SS, Jiang M, Kedaigle AJ, Arbez N, Marque LO, Eddings CR, Ranum PT, Whelan E, Tang A, Wang R, DeVine LR, Talbot CC, Cole RN, Ratovitski T, Davidson BL, Fraenkel E, Ross CA. Immortalized striatal precursor neurons from Huntington's disease patient-derived iPS cells as a platform for target identification and screening for experimental therapeutics. Hum Mol Genet 2021; 30:2469-2487. [PMID: 34296279 PMCID: PMC8643509 DOI: 10.1093/hmg/ddab200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/12/2022] Open
Abstract
We have previously established induced pluripotent stem cell (iPSC) models of Huntington's disease (HD), demonstrating CAG-repeat-expansion-dependent cell biological changes and toxicity. However, the current differentiation protocols are cumbersome and time consuming, making preparation of large quantities of cells for biochemical or screening assays difficult. Here, we report the generation of immortalized striatal precursor neurons (ISPNs) with normal (33) and expanded (180) CAG repeats from HD iPSCs, differentiated to a phenotype resembling medium spiny neurons (MSN), as a proof of principle for a more tractable patient-derived cell model. For immortalization, we used co-expression of the enzymatic component of telomerase hTERT and conditional expression of c-Myc. ISPNs can be propagated as stable adherent cell lines, and rapidly differentiated into highly homogeneous MSN-like cultures within 2 weeks, as demonstrated by immunocytochemical criteria. Differentiated ISPNs recapitulate major HD-related phenotypes of the parental iPSC model, including brain-derived neurotrophic factor (BDNF)-withdrawal-induced cell death that can be rescued by small molecules previously validated in the parental iPSC model. Proteome and RNA-seq analyses demonstrate separation of HD versus control samples by principal component analysis. We identified several networks, pathways, and upstream regulators, also found altered in HD iPSCs, other HD models, and HD patient samples. HD ISPN lines may be useful for studying HD-related cellular pathogenesis, and for use as a platform for HD target identification and screening experimental therapeutics. The described approach for generation of ISPNs from differentiated patient-derived iPSCs could be applied to a larger allelic series of HD cell lines, and to comparable modeling of other genetic disorders.
Collapse
Affiliation(s)
- Sergey S Akimov
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda J Kedaigle
- Department of Biological Engineering, Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Leonard O Marque
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chelsy R Eddings
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul T Ranum
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, The Raymond G Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emma Whelan
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Anthony Tang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ronald Wang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lauren R DeVine
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Conover C Talbot
- The Johns Hopkins School of Medicine, Institute for Basic Biomedical Sciences, Baltimore, MD 21205, USA
| | - Robert N Cole
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Beverly L Davidson
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, The Raymond G Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurology, Neuroscience and Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Picó S, Parras A, Santos-Galindo M, Pose-Utrilla J, Castro M, Fraga E, Hernández IH, Elorza A, Anta H, Wang N, Martí-Sánchez L, Belloc E, Garcia-Esparcia P, Garrido JJ, Ferrer I, Macías-García D, Mir P, Artuch R, Pérez B, Hernández F, Navarro P, López-Sendón JL, Iglesias T, Yang XW, Méndez R, Lucas JJ. CPEB alteration and aberrant transcriptome-polyadenylation lead to a treatable SLC19A3 deficiency in Huntington's disease. Sci Transl Med 2021; 13:eabe7104. [PMID: 34586830 DOI: 10.1126/scitranslmed.abe7104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sara Picó
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - Alberto Parras
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - María Santos-Galindo
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - Julia Pose-Utrilla
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid 28029, Spain
| | - Margarita Castro
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid 28049, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid,28029, Spain
| | - Enrique Fraga
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - Ivó H Hernández
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Facultad de Ciencias, Departamento de Biología (Unidad Docente Fisiología Animal), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ainara Elorza
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - Héctor Anta
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada I+D+i IMIM-IIBB (CSIC), Barcelona 08003, Spain.,Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Nan Wang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Laura Martí-Sánchez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid,28029, Spain.,Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona 08950, Spain
| | - Eulàlia Belloc
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Paula Garcia-Esparcia
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Institute of Neuropathology, IDIBELL-University Hospital Bellvitge, University of Barcelona, Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Juan J Garrido
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Department of Molecular, Cellular, and Developmental Neurobiology, Instituto Cajal (CSIC), Madrid 28002, Spain
| | - Isidro Ferrer
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Institute of Neuropathology, IDIBELL-University Hospital Bellvitge, University of Barcelona, Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Daniel Macías-García
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Pablo Mir
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Rafael Artuch
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid,28029, Spain.,Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona 08950, Spain
| | - Belén Pérez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid 28049, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid,28029, Spain
| | - Félix Hernández
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada I+D+i IMIM-IIBB (CSIC), Barcelona 08003, Spain.,Institute of Biomedical Research of Barcelona (IIBB-CSIC), Barcelona 08036, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - José Luis López-Sendón
- Department of Neurology, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Teresa Iglesias
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain.,Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid 28029, Spain
| | - X William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Raúl Méndez
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona 08028, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - José J Lucas
- Center for Molecular Biology "Severo Ochoa" (CBMSO) CSIC/UAM, Madrid, 28049, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, 28031, Spain
| |
Collapse
|
23
|
Peres DS, Theisen MC, Fialho MFP, Dalenogare DP, Rodrigues P, Kudsi SQ, Bernardes LDB, Ruviaro da Silva NA, Lückemeyer DD, Sampaio TB, Pereira GC, Mello FK, Ferreira J, Bochi GV, Oliveira SM, de David Antoniazzi CT, Trevisan G. TRPA1 involvement in depression- and anxiety-like behaviors in a progressive multiple sclerosis model in mice. Brain Res Bull 2021; 175:1-15. [PMID: 34280479 DOI: 10.1016/j.brainresbull.2021.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 06/25/2021] [Accepted: 07/14/2021] [Indexed: 11/28/2022]
Abstract
Progressive multiple sclerosis (PMS) is a neurological disease associated with the development of depression and anxiety, but treatments available are unsatisfactory. The transient receptor potential ankyrin 1 (TRPA1) is a cationic channel activated by reactive compounds, and the blockage of this receptor can reduce depression- and anxiety-like behaviors in naive mice. Thus, we investigated the role of TRPA1 in depression- and anxiety-like behaviors in a PMS model in mice. PMS model was induced in C57BL/6 female mice by the experimental autoimmune encephalomyelitis (EAE). Nine days after the PMS-EAE induction, behavioral tests (tail suspension and elevated plus maze tests) were performed to verify the effects of sertraline (positive control), selective TRPA1 antagonist (A-967,079), and antioxidants (α-lipoic acid and apocynin). The prefrontal cortex and hippocampus were collected to evaluate biochemical and inflammatory markers. PMS-EAE induction did not cause locomotor changes but triggered depression- and anxiety-like behaviors, which were reversed by sertraline, A-967,079, α-lipoic acid, or apocynin treatments. The neuroinflammatory markers (AIF1, GFAP, IL-1β, IL-17, and TNF-α) were increased in mice's hippocampus. Moreover, this model did not alter TRPA1 RNA expression levels in the hippocampus but decrease TRPA1 levels in the prefrontal cortex. Moreover, PMS-EAE induced an increase in NADPH oxidase and superoxide dismutase activities and TRPA1 endogenous agonist levels (hydrogen peroxide and 4-hydroxynonenal). TRPA1 plays a fundamental role in depression- and anxiety-like behaviors in a PMS-EAE model; thus, it could be a possible pharmacological target for treating these symptoms in PMS.
Collapse
Affiliation(s)
- Diulle Spat Peres
- Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | | | | | | | - Patrícia Rodrigues
- Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Sabrina Qader Kudsi
- Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | | | | | | | | | | | | | - Juliano Ferreira
- Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil
| | | | - Sara Marchesan Oliveira
- Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | | | - Gabriela Trevisan
- Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
24
|
Dubois C, Kong G, Tran H, Li S, Pang TY, Hannan AJ, Renoir T. Small Non-coding RNAs Are Dysregulated in Huntington's Disease Transgenic Mice Independently of the Therapeutic Effects of an Environmental Intervention. Mol Neurobiol 2021; 58:3308-3318. [PMID: 33675499 DOI: 10.1007/s12035-021-02342-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a trinucleotide repeat expansion in the huntingtin gene. Transcriptomic dysregulations are well-documented in HD and alterations in small non-coding RNAs (sncRNAs), particularly microRNAs (miRNAs), could underpin that phenomenon. Additionally, environmental enrichment (EE), which is used to model a stimulating lifestyle in pre-clinical research, has been shown to ameliorate HD-related symptoms. However, the mechanisms mediating the therapeutic effects of EE remain largely unknown. This study assessed the effect of EE on sncRNA expression in the striatum of female R6/1 transgenic HD mice at 12 weeks (prior to over motor deficits) and 20 weeks (fully symptomatic) of age. When comparing wild-type and R6/1 mice in the standard housing condition, we found 6 and 64 miRNAs that were differentially expressed at 12 and 20 weeks of age, respectively. The 6 miRNAs (miR-132, miR-212, miR-222, miR-1a, miR-467a, and miR-669c) were commonly dysregulated at both time points. Additionally, genotype had minor effects on the levels of other sncRNAs, in particular, 1 piRNA was dysregulated at 12 weeks of age, and at 20 weeks of age 11 piRNAs, 1 tRNA- and 2 snoRNA-derived fragments were altered in HD mice. No difference in the abundance of other sncRNA subtypes, including rRNA- and snRNA- derived fragments, were observed. While EE improved locomotor symptoms in HD, we found no effect of the housing condition on any of the sncRNA populations examined. Our findings show that HD mainly affects miRNAs and has a minor effect on other sncRNA populations. Furthermore, the therapeutic effects of EE are not associated with the rescue of these dysregulated sncRNAs and may therefore exert these experience-dependent effects via other molecular mechanisms.
Collapse
Affiliation(s)
- Celine Dubois
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia
| | - Geraldine Kong
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia
| | - Harvey Tran
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia
| | - Terence Y Pang
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia.
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, 3010, Australia.
| |
Collapse
|
25
|
Megret L, Gris B, Sasidharan Nair S, Cevost J, Wertz M, Aaronson J, Rosinski J, Vogt TF, Wilkinson H, Heiman M, Neri C. Shape deformation analysis reveals the temporal dynamics of cell-type-specific homeostatic and pathogenic responses to mutant huntingtin. eLife 2021; 10:64984. [PMID: 33618800 PMCID: PMC7901871 DOI: 10.7554/elife.64984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/31/2021] [Indexed: 01/06/2023] Open
Abstract
Loss of cellular homeostasis has been implicated in the etiology of several neurodegenerative diseases (NDs). However, the molecular mechanisms that underlie this loss remain poorly understood on a systems level in each case. Here, using a novel computational approach to integrate dimensional RNA-seq and in vivo neuron survival data, we map the temporal dynamics of homeostatic and pathogenic responses in four striatal cell types of Huntington’s disease (HD) model mice. This map shows that most pathogenic responses are mitigated and most homeostatic responses are decreased over time, suggesting that neuronal death in HD is primarily driven by the loss of homeostatic responses. Moreover, different cell types may lose similar homeostatic processes, for example, endosome biogenesis and mitochondrial quality control in Drd1-expressing neurons and astrocytes. HD relevance is validated by human stem cell, genome-wide association study, and post-mortem brain data. These findings provide a new paradigm and framework for therapeutic discovery in HD and other NDs.
Collapse
Affiliation(s)
- Lucile Megret
- Sorbonne Université, Centre National de la Recherche Scientifique UMR 8256, INSERM ERL U1164, Paris, France
| | - Barbara Gris
- Sorbonne Université, Centre National de la Recherche Scientifique, Laboratoire Jacques-Louis Lyons (LJLL), Paris, France
| | - Satish Sasidharan Nair
- Sorbonne Université, Centre National de la Recherche Scientifique UMR 8256, INSERM ERL U1164, Paris, France
| | - Jasmin Cevost
- Sorbonne Université, Centre National de la Recherche Scientifique UMR 8256, INSERM ERL U1164, Paris, France
| | - Mary Wertz
- MIT, Broad Institute, MIT, Picower Institute for Learning and Memory, Cambridge, United States
| | | | | | | | | | - Myriam Heiman
- MIT, Broad Institute, MIT, Picower Institute for Learning and Memory, Cambridge, United States
| | - Christian Neri
- Sorbonne Université, Centre National de la Recherche Scientifique UMR 8256, INSERM ERL U1164, Paris, France
| |
Collapse
|
26
|
Dual Profile of Environmental Enrichment and Autistic-Like Behaviors in the Maternal Separated Model in Rats. Int J Mol Sci 2021; 22:ijms22031173. [PMID: 33503967 PMCID: PMC7865216 DOI: 10.3390/ijms22031173] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Environmental Enrichment (EE) has been suggested as a possible therapeutic intervention for neurodevelopmental disorders such as autism. Although the benefits of this therapeutic method have been reported in some animal models and human studies, the unknown pathophysiology of autism as well as number of conflicting results, urge for further examination of the therapeutic potential of EE in autism. Therefore, the aim of this study was to examine the effects of environmental enrichment on autism-related behaviors which were induced in the maternal separation (MS) animal model. MATERIAL AND METHODS Maternally separated (post-natal day (PND) 1-14, 3h/day) and control male rats were at weaning (PND21) age equally divided into rats housed in enriched environment and normal environment. At adolescence (PND42-50), the four groups were behaviorally tested for direct social interaction, sociability, repetitive behaviors, anxiety behavior, and locomotion. Following completion of the behavioral tests, the blood and brain tissue samples were harvested in order to assess plasma level of brain derived neurotrophic factor (BDNF) and structural plasticity of brain using ELISA and stereological methods respectively. RESULTS We found that environmental enrichment reduced repetitive behaviors but failed to improve the impaired sociability and anxiety behaviors which were induced by maternal separation. Indeed, EE exacerbated anxiety and social behaviors deficits in association with increased plasma BDNF level, larger volume of the hippocampus and infra-limbic region and higher number of neurons in the infra-limbic area (p < 0.05). Conclusion: We conclude that environmental enrichment has a significant improvement effect on the repetitive behavior as one of the core autistic-like behaviors induced by maternal separation but has negative effect on the anxiety and social behaviors which might have been modulated by BDNF.
Collapse
|
27
|
Expanding genes, repeating themes and therapeutic schemes: The neurobiology of tandem repeat disorders. Neurobiol Dis 2020; 144:105053. [PMID: 32810583 DOI: 10.1016/j.nbd.2020.105053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
28
|
Cheong RY, Gabery S, Petersén Å. The Role of Hypothalamic Pathology for Non-Motor Features of Huntington's Disease. J Huntingtons Dis 2020; 8:375-391. [PMID: 31594240 PMCID: PMC6839491 DOI: 10.3233/jhd-190372] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Huntington’s disease (HD) is a fatal genetic neurodegenerative disorder. It has mainly been considered a movement disorder with cognitive symptoms and these features have been associated with pathology of the striatum and cerebral cortex. Importantly, individuals with the mutant huntingtin gene suffer from a spectrum of non-motor features often decades before the motor disorder manifests. These symptoms and signs include a range of psychiatric symptoms, sleep problems and metabolic changes with weight loss particularly in later stages. A higher body mass index at diagnosis is associated with slower disease progression. The common psychiatric symptom of apathy progresses with the disease. The fact that non-motor features are present early in the disease and that they show an association to disease progression suggest that unravelling the underlying neurobiological mechanisms may uncover novel targets for early disease intervention and better symptomatic treatment. The hypothalamus and the limbic system are important brain regions that regulate emotion, social cognition, sleep and metabolism. A number of studies using neuroimaging, postmortem human tissue and genetic manipulation in animal models of the disease has collectively shown that the hypothalamus and the limbic system are affected in HD. These findings include the loss of neuropeptide-expressing neurons such as orexin (hypocretin), oxytocin, vasopressin, somatostatin and VIP, and increased levels of SIRT1 in distinct nuclei of the hypothalamus. This review provides a summary of the results obtained so far and highlights the potential importance of these changes for the understanding of non-motor features in HD.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
29
|
Gubert C, Renoir T, Hannan AJ. Why Woody got the blues: The neurobiology of depression in Huntington's disease. Neurobiol Dis 2020; 142:104958. [PMID: 32526274 DOI: 10.1016/j.nbd.2020.104958] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/02/2020] [Accepted: 06/03/2020] [Indexed: 02/03/2023] Open
Abstract
Huntington's disease (HD) is an extraordinary disorder that usually strikes when individuals are in the prime of their lives, as was the case for the influential 20th century musician Woody Guthrie. HD demonstrates the exceptionally fine line between life and death in such 'genetic diseases', as the only difference between those who suffer horribly and die slowly of this disease is often just a handful of extra tandem repeats (beyond the normal polymorphic range) in a genome that constitutes over 3 billion paired nucleotides of DNA. Furthermore, HD presents as a complex and heterogenous combination of psychiatric, cognitive and motor symptoms, so can appear as an unholy trinity of 'three disorders in one'. The autosomal dominant nature of the disorder is also extremely challenging for affected families, as a 'flip of a coin' dictates which children inherit the mutation from their affected parent, and the gene-negative family members bear the burden of caring for the other half of the family that is affected. In this review, we will focus on one of the earliest, and most devastating, symptoms associated with HD, depression, which has been reported to affect approximately half of gene-positive HD family members. We will discuss the pathogenesis of HD, and depressive symptoms in particular, including molecular and cellular mechanisms, and potential genetic and environmental modifiers. This expanding understanding of HD pathogenesis may not only lead to novel therapeutic options for HD families, but may also provide insights into depression in the wider population, which has the greatest burden of disease of any disorder and an enormous unmet need for new therapies.
Collapse
Affiliation(s)
- Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
30
|
Schachtschneider KM, Welge ME, Auvil LS, Chaki S, Rund LA, Madsen O, Elmore MR, Johnson RW, Groenen MA, Schook LB. Altered Hippocampal Epigenetic Regulation Underlying Reduced Cognitive Development in Response to Early Life Environmental Insults. Genes (Basel) 2020; 11:genes11020162. [PMID: 32033187 PMCID: PMC7074491 DOI: 10.3390/genes11020162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is involved in learning and memory and undergoes significant growth and maturation during the neonatal period. Environmental insults during this developmental timeframe can have lasting effects on brain structure and function. This study assessed hippocampal DNA methylation and gene transcription from two independent studies reporting reduced cognitive development stemming from early life environmental insults (iron deficiency and porcine reproductive and respiratory syndrome virus (PRRSv) infection) using porcine biomedical models. In total, 420 differentially expressed genes (DEGs) were identified between the reduced cognition and control groups, including genes involved in neurodevelopment and function. Gene ontology (GO) terms enriched for DEGs were associated with immune responses, angiogenesis, and cellular development. In addition, 116 differentially methylated regions (DMRs) were identified, which overlapped 125 genes. While no GO terms were enriched for genes overlapping DMRs, many of these genes are known to be involved in neurodevelopment and function, angiogenesis, and immunity. The observed altered methylation and expression of genes involved in neurological function suggest reduced cognition in response to early life environmental insults is due to altered cholinergic signaling and calcium regulation. Finally, two DMRs overlapped with two DEGs, VWF and LRRC32, which are associated with blood brain barrier permeability and regulatory T-cell activation, respectively. These results support the role of altered hippocampal DNA methylation and gene expression in early life environmentally-induced reductions in cognitive development across independent studies.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Michael E. Welge
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Loretta S. Auvil
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Sulalita Chaki
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Monica R.P. Elmore
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Martien A.M. Groenen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
- Correspondence:
| |
Collapse
|
31
|
Comella Bolla A, Valente T, Miguez A, Brito V, Gines S, Solà C, Straccia M, Canals JM. CD200 is up-regulated in R6/1 transgenic mouse model of Huntington's disease. PLoS One 2019; 14:e0224901. [PMID: 31790427 PMCID: PMC6886799 DOI: 10.1371/journal.pone.0224901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/23/2019] [Indexed: 01/17/2023] Open
Abstract
In Huntington's disease (HD), striatal medium spiny neurons (MSNs) are particularly sensitive to the presence of a CAG repeat in the huntingtin (HTT) gene. However, there are many evidences that cells from the peripheral immune system and central nervous system (CNS) immune cells, namely microglia, play an important role in the etiology and the progression of HD. However, it remains unclear whether MSNs neurodegeneration is mediated by a non-cell autonomous mechanism. The homeostasis in the healthy CNS is maintained by several mechanisms of interaction between all brain cells. Neurons can control microglia activation through several inhibitory mechanisms, such as the CD200-CD200R1 interaction. Due to the complete lack of knowledge about the CD200-CD200R1 system in HD, we determined the temporal patterns of CD200 and CD200R1 expression in the neocortex, hippocampus and striatum in the HD mouse models R6/1 and HdhQ111/7 from pre-symptomatic to manifest stages. In order to explore any alteration in the peripheral immune system, we also studied the levels of expression of CD200 and CD200R1 in whole blood. Although CD200R1 expression was not altered, we observed and increase in CD200 gene expression and protein levels in the brain parenchyma of all the regions we examined, along with HD pathogenesis in R6/1 mice. Interestingly, the expression of CD200 mRNA was also up-regulated in blood following a similar temporal pattern. These results suggest that canonical neuronal-microglial communication through CD200-CD200R1 interaction is not compromised, and CD200 up-regulation in R6/1 brain parenchyma could represent a neurotrophic signal to sustain or extend neuronal function in the latest stages of HD as pro-survival mechanism.
Collapse
Affiliation(s)
- Andrea Comella Bolla
- Stem Cells and Regenerative Medicine Laboratory, Production and Validation Center of Advanced Therapies (Creatio), Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Tony Valente
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona–Consejo Superior de Investigaciones Científicas (IIBB–CSIC), Barcelona, Spain
| | - Andres Miguez
- Stem Cells and Regenerative Medicine Laboratory, Production and Validation Center of Advanced Therapies (Creatio), Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Veronica Brito
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Silvia Gines
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Carme Solà
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Cerebral Ischemia and Neurodegeneration, Institut d’Investigacions Biomèdiques de Barcelona–Consejo Superior de Investigaciones Científicas (IIBB–CSIC), Barcelona, Spain
| | - Marco Straccia
- Stem Cells and Regenerative Medicine Laboratory, Production and Validation Center of Advanced Therapies (Creatio), Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Josep M. Canals
- Stem Cells and Regenerative Medicine Laboratory, Production and Validation Center of Advanced Therapies (Creatio), Department of Biomedicine, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Neuroscience Institute, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
32
|
Therapeutic efficacy of environmental enrichment for substance use disorders. Pharmacol Biochem Behav 2019; 188:172829. [PMID: 31778722 DOI: 10.1016/j.pbb.2019.172829] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022]
Abstract
Addiction to drug and alcohol is regarded as a major health problem worldwide for which available treatments show limited effectiveness. The biggest challenge remains to enhance the capacities of interventions to reduce craving, prevent relapse and promote long-term recovery. New strategies to meet these challenges are being explored. Findings from preclinical work suggest that environmental enrichment (EE) holds therapeutic potential for the treatment of substance use disorders, as demonstrated in a number of animal models of drug abuse. The EE intervention introduced after drug exposure leads to attenuation of compulsive drug taking, attenuation of the rewarding (and reinforcing) effects of drugs, reductions in control of behavior by drug cues, and, very importantly, relapse prevention. Clinical work also suggests that multidimensional EE interventions (involving physical activity, social interaction, vocational training, recreational and community involvement) might produce similar therapeutic effects, if implemented continuously and rigorously. In this review we survey preclinical and clinical studies assessing the efficacy of EE as a behavioral intervention for substance use disorders and address related challenges. We also review work providing empirical evidence for EE-induced neuroplasticity within the mesocorticolimbic system that is believed to contribute to the seemingly therapeutic effects of EE on drug and alcohol-related behaviors.
Collapse
|
33
|
Campbell EJ, Jin S, Lawrence AJ. Environmental enrichment reduces the propensity to relapse following punishment-imposed abstinence of alcohol seeking. Physiol Behav 2019; 210:112638. [DOI: 10.1016/j.physbeh.2019.112638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/11/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023]
|
34
|
Short-term environmental enrichment, and not physical exercise, alleviate cognitive decline and anxiety from middle age onwards without affecting hippocampal gene expression. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2019; 19:1143-1169. [DOI: 10.3758/s13415-019-00743-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
35
|
Gubert C, Hannan AJ. Environmental enrichment as an experience-dependent modulator of social plasticity and cognition. Brain Res 2019; 1717:1-14. [DOI: 10.1016/j.brainres.2019.03.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/11/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
|
36
|
Tobore TO. Towards a comprehensive understanding of the contributions of mitochondrial dysfunction and oxidative stress in the pathogenesis and pathophysiology of Huntington's disease. J Neurosci Res 2019; 97:1455-1468. [DOI: 10.1002/jnr.24492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022]
|
37
|
Galts CP, Bettio LE, Jewett DC, Yang CC, Brocardo PS, Rodrigues ALS, Thacker JS, Gil-Mohapel J. Depression in neurodegenerative diseases: Common mechanisms and current treatment options. Neurosci Biobehav Rev 2019; 102:56-84. [DOI: 10.1016/j.neubiorev.2019.04.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 04/02/2019] [Indexed: 12/19/2022]
|
38
|
Franco NH, Olsson IAS. “How Sick Must Your Mouse Be?” — An Analysis of the Use of Animal Models in Huntington's Disease Research. Altern Lab Anim 2019; 40:271-83. [DOI: 10.1177/026119291204000506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Nuno H. Franco
- IBMC — Institute for Molecular and Cell Biology (Laboratory Animal Science Group), University of Porto, Porto, Portugal
| | - I. Anna S. Olsson
- IBMC — Institute for Molecular and Cell Biology (Laboratory Animal Science Group), University of Porto, Porto, Portugal
| |
Collapse
|
39
|
Spatial memory in Huntington’s disease: A comparative review of human and animal data. Neurosci Biobehav Rev 2019; 98:194-207. [DOI: 10.1016/j.neubiorev.2019.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 12/24/2022]
|
40
|
Zajac MS, Renoir T, Perreau VM, Li S, Adams W, van den Buuse M, Hannan AJ. Short-Term Environmental Stimulation Spatiotemporally Modulates Specific Serotonin Receptor Gene Expression and Behavioral Pharmacology in a Sexually Dimorphic Manner in Huntington's Disease Transgenic Mice. Front Mol Neurosci 2018; 11:433. [PMID: 30618600 PMCID: PMC6295568 DOI: 10.3389/fnmol.2018.00433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/08/2018] [Indexed: 11/29/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a tandem repeat mutation encoding an expanded polyglutamine tract in the huntingtin protein, which leads to cognitive, psychiatric and motor dysfunction. Exposure to environmental enrichment (EE), which enhances levels of cognitive stimulation and physical activity, has therapeutic effects on cognitive, affective and motor function of transgenic HD mice. The present study investigated gene expression changes and behavioral pharmacology in male and female R6/1 transgenic HD mice at an early time-point in HD progression associated with onset of cognitive and affective abnormalities, following EE and exercise (wheel running) interventions. We have demonstrated changes in expression levels of the serotonin (5-HT) receptor Htr1a, Htr1b, Htr2a and Htr2c genes (encoding the 5-HT1A, 5-HT1B, 5-HT2A and 5-HT2C receptors, respectively) in HD brains at 8 weeks of age, using quantitative real-time PCR. In contrast, expression of the serotonin transporter (SerT, also known as 5-HTT or Slc6a4) was not altered in these brains. Furthermore, we identified region-specific, sex-specific and environmentally regulated (comparing EE, exercise and standard housing conditions) impacts on gene expression of particular 5-HT receptors, as well as SerT. For example, SerT gene expression was upregulated by exercise (wheel running from 6 to 8 weeks of age) in the hippocampus. Interestingly, when EE was introduced from 6 to 8 weeks of age, Htr2a gene expression was upregulated in the cortex, striatum and hippocampus of male mice. EE also rescued the functional activity of 5-HT2 receptors as observed in the head-twitch test, reflecting sexually dimorphic effects of environmental stimulation. These findings demonstrate that disruption of the serotonergic system occurs early in HD pathogenesis and, together with previous findings, show that the timing and duration of environmental interventions are critical in terms of their ability to modify gene expression. This study is the first to show that EE is able to selectively enhance both gene expression of a neurotransmitter receptor and the functional consequences on behavioral pharmacology, and links this molecular modulation to the therapeutic effects of environmental stimulation in this neurodegenerative disease.
Collapse
Affiliation(s)
- Michelle S Zajac
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Victoria M Perreau
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Wendy Adams
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Maarten van den Buuse
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
41
|
Molecular Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:289-333. [PMID: 30409256 DOI: 10.1016/bs.irn.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is a rare monogenic neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in the huntingtin gene resulting in the formation of intranuclear inclusions of mutated huntingtin. The accumulation of mutated huntingtin leads to loss of GABAergic medium spiny neurons (MSNs); subsequently resulting in the development of chorea, cognitive dysfunction and psychiatric symptoms. Premanifest HD gene expansion carriers, provide a unique cohort to examine very early molecular changes, occurring before the development of overt symptoms, to elucidate disease pathophysiology and identify reliable biomarkers of HD progression. Positron emission tomography (PET) is a non-invasive molecular imaging technique allowing the evaluation of specific molecular targets in vivo. Selective PET radioligands provide invaluable tools to investigate the role of the dopaminergic system, brain metabolism, microglial activation, phosphodiesterase 10A, and cannabinoid, GABA, adenosine and opioid receptors in HD. PET has been employed to monitor disease progression aiming to identify a reliable biomarker to predict phenoconversion from premanifest to manifest HD.
Collapse
|
42
|
Antidepressant Effects of Probucol on Early-Symptomatic YAC128 Transgenic Mice for Huntington's Disease. Neural Plast 2018; 2018:4056383. [PMID: 30186318 PMCID: PMC6112232 DOI: 10.1155/2018/4056383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/28/2018] [Accepted: 07/26/2018] [Indexed: 11/17/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a trinucleotide expansion in the HD gene, resulting in an extended polyglutamine tract in the protein huntingtin. HD is traditionally viewed as a movement disorder, but cognitive and neuropsychiatric symptoms also contribute to the clinical presentation. Depression is one of the most common psychiatric disturbances in HD, present even before manifestation of motor symptoms. Diagnosis and treatment of depression in HD-affected individuals are essential aspects of clinical management in this population, especially owing to the high risk of suicide. This study investigated whether chronic administration of the antioxidant probucol improved motor and affective symptoms as well as hippocampal neurogenic function in the YAC128 transgenic mouse model of HD during the early- to mild-symptomatic stages of disease progression. The motor performance and affective symptoms were monitored using well-validated behavioral tests in YAC128 mice and age-matched wild-type littermates at 2, 4, and 6 months of age, after 1, 3, or 5 months of treatment with probucol (30 mg/kg/day via water supplementation, starting on postnatal day 30). Endogenous markers were used to assess the effect of probucol on cell proliferation (Ki-67 and proliferation cell nuclear antigen (PCNA)) and neuronal differentiation (doublecortin (DCX)) in the hippocampal dentate gyrus (DG). Chronic treatment with probucol reduced the occurrence of depressive-like behaviors in early- and mild-symptomatic YAC128 mice. Functional improvements were not accompanied by increased progenitor cell proliferation and neuronal differentiation. Our findings provide evidence that administration of probucol may be of clinical benefit in the management of early- to mild-symptomatic HD.
Collapse
|
43
|
Novati A, Hentrich T, Wassouf Z, Weber JJ, Yu-Taeger L, Déglon N, Nguyen HP, Schulze-Hentrich JM. Environment-dependent striatal gene expression in the BACHD rat model for Huntington disease. Sci Rep 2018; 8:5803. [PMID: 29643462 PMCID: PMC5895842 DOI: 10.1038/s41598-018-24243-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutation in the huntingtin (HTT) gene which results in progressive neurodegeneration in the striatum, cortex, and eventually most brain areas. Despite being a monogenic disorder, environmental factors influence HD characteristics. Both human and mouse studies suggest that mutant HTT (mHTT) leads to gene expression changes that harbor potential to be modulated by the environment. Yet, the underlying mechanisms integrating environmental cues into the gene regulatory program have remained largely unclear. To better understand gene-environment interactions in the context of mHTT, we employed RNA-seq to examine effects of maternal separation (MS) and environmental enrichment (EE) on striatal gene expression during development of BACHD rats. We integrated our results with striatal consensus modules defined on HTT-CAG length and age-dependent co-expression gene networks to relate the environmental factors with disease progression. While mHTT was the main determinant of expression changes, both MS and EE were capable of modulating these disturbances, resulting in distinctive and in several cases opposing effects of MS and EE on consensus modules. This bivalent response to maternal separation and environmental enrichment may aid in explaining their distinct effects observed on disease phenotypes in animal models of HD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Arianna Novati
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Thomas Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Zinah Wassouf
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Jonasz J Weber
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Nicole Déglon
- Department of Clinical Neurosciences (DNC), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany. .,Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany.
| | | |
Collapse
|
44
|
Novel approaches to alcohol rehabilitation: Modification of stress-responsive brain regions through environmental enrichment. Neuropharmacology 2018; 145:25-36. [PMID: 29477298 DOI: 10.1016/j.neuropharm.2018.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/05/2018] [Accepted: 02/21/2018] [Indexed: 01/08/2023]
Abstract
Relapse remains the most prominent hurdle to successful rehabilitation from alcoholism. The neural mechanisms underlying relapse are complex, but our understanding of the brain regions involved, the anatomical circuitry and the modulation of specific nuclei in the context of stress and cue-induced relapse have improved significantly in recent years. In particular, stress is now recognised as a significant trigger for relapse, adding to the well-established impact of chronic stress to escalate alcohol consumption. It is therefore unsurprising that the stress-responsive regions of the brain have also been implicated in alcohol relapse, such as the nucleus accumbens, amygdala and the hypothalamus. Environmental enrichment is a robust experimental paradigm which provides a non-pharmacological tool to alter stress response and, separately, alcohol-seeking behaviour and symptoms of withdrawal. In this review, we examine and consolidate the preclinical evidence that alcohol seeking behaviour and stress-induced relapse are modulated by environmental enrichment, and these are primarily mediated by modification of neural activity within the key nodes of the addiction circuitry. Finally, we discuss the limited clinical evidence that stress-reducing approaches such as mindfulness could potentially serve as adjunctive therapy in the treatment of alcoholism. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
|
45
|
Sampedro-Piquero P, Begega A. Environmental Enrichment as a Positive Behavioral Intervention Across the Lifespan. Curr Neuropharmacol 2018; 15:459-470. [PMID: 27012955 PMCID: PMC5543669 DOI: 10.2174/1570159x14666160325115909] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/30/2015] [Accepted: 03/16/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In recent decades, the interest in behavioral interventions has been growing due to the higher prevalence of age-related cognitive impairments. Hence, behavioral interventions, such as cognitive stimulation and physical activity, and along with these, our lifestyle (education level, work position, frequency of cognitive and social activities) have shown important benefits during the cognitive impairment, dementia and even recovery after brain injury. This is due to the fact that this type of intervention and activities promote the formation of a cognitive and brain reserve that allows tolerating brain damage during a long period of time without the appearance of cognitive symptoms. With regard to this, animal models have proved very useful in providing information about the brain mechanisms involved in the development of these cognitive and brain reserves and how they interact with each other. METHODS We summarize several studies showing the positive effects of Environmental Enrichment (EE), understood as a housing condition in which animals benefit from the sensory, physical, cognitive and social stimulation provided, on brain and cognitive functions usually impaired during aging. RESULTS Most of studies have shown that EE is a successful protocol to improve cognitive functions and reduce anxiety-related behaviors across the lifespan, as well as in animal models of neurodegenerative diseases. CONCLUSION Therefore, EE is a laboratory condition in which some aspects of an active lifestyle are reproduced.
Collapse
Affiliation(s)
- P Sampedro-Piquero
- Department of Biological and Health Psychology, Autonomous University of Madrid, Cantoblanco 28049, Madrid, Spain
| | - A Begega
- Neuroscience Laboratory, Psychology Department, University of Oviedo, Plaza Feijoo s/n 33003 Oviedo, INEUROPA, Spain
| |
Collapse
|
46
|
Rogers J, Renoir T, Hannan AJ. Gene-environment interactions informing therapeutic approaches to cognitive and affective disorders. Neuropharmacology 2017; 145:37-48. [PMID: 29277490 DOI: 10.1016/j.neuropharm.2017.12.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023]
Abstract
Gene-environment interactions drive experience-dependent changes in the brain that alter cognition, emotion and behaviour. Positive engagement with the environment, through novel experience and physical activity, can improve brain function, although the mechanisms mediating such experience-dependent plasticity remain to be fully elucidated. In this article, we discuss the therapeutic value of environmental stimuli, exercise and environmental enrichment (EE), for cognitive and affective disorders, with implications for the understanding and treatment of depression and anxiety disorders. We demonstrate that environmental manipulations are potential therapeutic strategies for improving outcomes in these psychiatric disorders, including beneficial impacts on cognition. We discuss how EE and exercise are therapeutic environmental interventions impacting both affective and cognitive function. Serotonergic (5-HTergic) signaling is strongly implicated in the manifestation of psychiatric disorders and regulates cognitive and emotional processing that can underpin them. Thus, we focus on evidence implicating the serotonergic system in mediating gene-environment interactions to EE and exercise. Finally, we discuss robust gene-environment interactions associated with EE and exercise interventions, and their impacts on specific brain areas, particularly the hippocampus. We focus on potential mediators of this experience-dependent plasticity, including adult neurogenesis and brain-derived neurotrophic factor (BDNF). Furthermore, we explore molecular and cellular mechanisms of experience-dependent plasticity that potentially underlie the restoration of affective and cognitive phenotypes, thus identifying novel therapeutic targets. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- Jake Rogers
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia.
| |
Collapse
|
47
|
Ouk K, Aungier J, Cuesta M, Morton AJ. Chronic paroxetine treatment prevents disruption of methamphetamine-sensitive circadian oscillator in a transgenic mouse model of Huntington's disease. Neuropharmacology 2017; 131:337-350. [PMID: 29274752 DOI: 10.1016/j.neuropharm.2017.12.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/31/2017] [Accepted: 12/19/2017] [Indexed: 02/04/2023]
Abstract
Circadian abnormalities seen in Huntington's disease (HD) patients are recapitulated in several HD transgenic mouse models. In mice, alongside the master clock located in the suprachiasmatic nucleus (SCN), two other oscillators may influence circadian behaviour. These are the food-entrainable oscillator (FEO) and the methamphetamine-sensitive circadian oscillator (MASCO). SCN- and MASCO- (but not FEO-) driven rhythms are progressively disrupted in the R6/2 mouse model of HD. MASCO-driven rhythms are induced by chronic treatment with low dose of methamphetamine and characterised by an increase in period length to greater than 24 h. Interestingly, the rhythms mediated by MASCO deteriorate earlier than those mediated by the SCN in R6/2 mice. Here, we used a pharmacological strategy to investigate the mechanisms underlying MASCO-driven rhythms in WT mice. In contrast to methamphetamine, chronic cocaine was ineffective in generating a MASCO-like component of activity although it markedly increased locomotion. Furthermore, neither blocking dopamine (DA) receptors (with the DA antagonist haloperidol) nor blocking neurotransmission by inhibiting the activity of vesicular monoamine transporter (with reserpine) prevented the expression of the MASCO-driven rhythms, although both treatments downregulated locomotor activity. Interestingly, chronic treatment with paroxetine, a serotonin-specific reuptake inhibitor commonly used as antidepressant in HD, was able to restore the expression of MASCO-driven rhythms in R6/2 mice. Thus, MASCO-driven rhythms appear to be mediated by both serotoninergic and dopaminergic systems. This supports the idea that abnormalities in MASCO output may contribute to both the HD circadian and psychiatric phenotype.
Collapse
Affiliation(s)
- Koliane Ouk
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, United Kingdom
| | - Juliet Aungier
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, United Kingdom
| | - Marc Cuesta
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, United Kingdom
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, United Kingdom.
| |
Collapse
|
48
|
Gardiner SL, van Belzen MJ, Boogaard MW, van Roon-Mom WMC, Rozing MP, van Hemert AM, Smit JH, Beekman ATF, van Grootheest G, Schoevers RA, Oude Voshaar RC, Roos RAC, Comijs HC, Penninx BWJH, van der Mast RC, Aziz NA. Huntingtin gene repeat size variations affect risk of lifetime depression. Transl Psychiatry 2017; 7:1277. [PMID: 29225330 PMCID: PMC5802693 DOI: 10.1038/s41398-017-0042-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/29/2017] [Accepted: 09/15/2017] [Indexed: 11/23/2022] Open
Abstract
Huntington disease (HD) is a severe neuropsychiatric disorder caused by a cytosine-adenine-guanine (CAG) repeat expansion in the HTT gene. Although HD is frequently complicated by depression, it is still unknown to what extent common HTT CAG repeat size variations in the normal range could affect depression risk in the general population. Using binary logistic regression, we assessed the association between HTT CAG repeat size and depression risk in two well-characterized Dutch cohorts─the Netherlands Study of Depression and Anxiety and the Netherlands Study of Depression in Older Persons─including 2165 depressed and 1058 non-depressed persons. In both cohorts, separately as well as combined, there was a significant non-linear association between the risk of lifetime depression and HTT CAG repeat size in which both relatively short and relatively large alleles were associated with an increased risk of depression (β = -0.292 and β = 0.006 for the linear and the quadratic term, respectively; both P < 0.01 after adjustment for the effects of sex, age, and education level). The odds of lifetime depression were lowest in persons with a HTT CAG repeat size of 21 (odds ratio: 0.71, 95% confidence interval: 0.52 to 0.98) compared to the average odds in the total cohort. In conclusion, lifetime depression risk was higher with both relatively short and relatively large HTT CAG repeat sizes in the normal range. Our study provides important proof-of-principle that repeat polymorphisms can act as hitherto unappreciated but complex genetic modifiers of depression.
Collapse
Affiliation(s)
- Sarah L. Gardiner
- 0000000089452978grid.10419.3dDepartments of Neurology, Leiden University Medical Centre, Leiden, The Netherlands ,0000000089452978grid.10419.3dDepartments of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Martine J. van Belzen
- 0000000089452978grid.10419.3dDepartments of Clinical Genetics, and Leiden University Medical Centre, Leiden, The Netherlands
| | - Merel W. Boogaard
- 0000000089452978grid.10419.3dDepartments of Neurology, Leiden University Medical Centre, Leiden, The Netherlands ,0000000089452978grid.10419.3dDepartments of Clinical Genetics, and Leiden University Medical Centre, Leiden, The Netherlands
| | - Willeke M. C. van Roon-Mom
- 0000000089452978grid.10419.3dDepartments of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maarten P. Rozing
- 0000 0001 0674 042Xgrid.5254.6Department of Public Health, Section of Social Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Albert M. van Hemert
- 0000000089452978grid.10419.3dDepartments of Psychiatry, Leiden University Medical Centre, Leiden, The Netherlands
| | - Johannes H. Smit
- 0000 0001 0686 3219grid.466632.3Department of Psychiatry, and EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Aartjan T. F. Beekman
- 0000 0001 0686 3219grid.466632.3Department of Psychiatry, and EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Gerard van Grootheest
- 0000 0001 0686 3219grid.466632.3Department of Psychiatry, and EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Robert A. Schoevers
- 0000 0000 9558 4598grid.4494.dDepartment of Psychiatry, University Medical Centre Groningen, Groningen, The Netherlands
| | - Richard C. Oude Voshaar
- 0000 0000 9558 4598grid.4494.dDepartment of Psychiatry, University Medical Centre Groningen, Groningen, The Netherlands
| | - Raymund A. C. Roos
- 0000000089452978grid.10419.3dDepartments of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hannie C. Comijs
- 0000 0001 0686 3219grid.466632.3Department of Psychiatry, and EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Brenda W. J. H. Penninx
- 0000 0001 0686 3219grid.466632.3Department of Psychiatry, and EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Roos C. van der Mast
- 0000000089452978grid.10419.3dDepartments of Psychiatry, Leiden University Medical Centre, Leiden, The Netherlands ,0000 0001 0790 3681grid.5284.b Department of Psychiatry, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - N. Ahmad Aziz
- 0000000089452978grid.10419.3dDepartments of Neurology, Leiden University Medical Centre, Leiden, The Netherlands ,0000000121901201grid.83440.3bDepartment of Neurodegenerative Disease, UCL Huntington’s Disease Centre, University College London Institute of Neurology, London, United Kingdom
| |
Collapse
|
49
|
Lamirault C, Yu-Taeger L, Doyère V, Riess O, Nguyen HP, El Massioui N. Altered reactivity of central amygdala to GABA A R antagonist in the BACHD rat model of Huntington disease. Neuropharmacology 2017; 123:136-147. [DOI: 10.1016/j.neuropharm.2017.05.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/05/2017] [Accepted: 05/30/2017] [Indexed: 11/16/2022]
|
50
|
Rogers J, Li S, Lanfumey L, Hannan AJ, Renoir T. Environmental enrichment reduces innate anxiety with no effect on depression-like behaviour in mice lacking the serotonin transporter. Behav Brain Res 2017. [DOI: 10.1016/j.bbr.2017.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|