1
|
Kong L, Chen Y, Shen Y, Zhang D, Wei C, Lai J, Hu S. Progress and Implications from Genetic Studies of Bipolar Disorder. Neurosci Bull 2024; 40:1160-1172. [PMID: 38206551 PMCID: PMC11306703 DOI: 10.1007/s12264-023-01169-9] [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: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 01/12/2024] Open
Abstract
With the advancements in gene sequencing technologies, including genome-wide association studies, polygenetic risk scores, and high-throughput sequencing, there has been a tremendous advantage in mapping a detailed blueprint for the genetic model of bipolar disorder (BD). To date, intriguing genetic clues have been identified to explain the development of BD, as well as the genetic association that might be applied for the development of susceptibility prediction and pharmacogenetic intervention. Risk genes of BD, such as CACNA1C, ANK3, TRANK1, and CLOCK, have been found to be involved in various pathophysiological processes correlated with BD. Although the specific roles of these genes have yet to be determined, genetic research on BD will help improve the prevention, therapeutics, and prognosis in clinical practice. The latest preclinical and clinical studies, and reviews of the genetics of BD, are analyzed in this review, aiming to summarize the progress in this intriguing field and to provide perspectives for individualized, precise, and effective clinical practice.
Collapse
Affiliation(s)
- Lingzhuo Kong
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yiqing Chen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuting Shen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Wei
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianbo Lai
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Shaohua Hu
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
2
|
Hughes DN, Klein MH, Walder-Christensen KK, Thomas GE, Grossman Y, Waters D, Matthews AE, Carson WE, Filali Y, Tsyglakova M, Fink A, Gallagher NM, Perez-Balaguer M, McClung CA, Zarate JM, Hultman RC, Mague SD, Carlson DE, Dzirasa K. A widespread electrical brain network encodes anxiety in health and depressive states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600900. [PMID: 38979139 PMCID: PMC11230447 DOI: 10.1101/2024.06.26.600900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In rodents, anxiety is charactered by heightened vigilance during low-threat and uncertain situations. Though activity in the frontal cortex and limbic system are fundamental to supporting this internal state, the underlying network architecture that integrates activity across brain regions to encode anxiety across animals and paradigms remains unclear. Here, we utilize parallel electrical recordings in freely behaving mice, translational paradigms known to induce anxiety, and machine learning to discover a multi-region network that encodes the anxious brain-state. The network is composed of circuits widely implicated in anxiety behavior, it generalizes across many behavioral contexts that induce anxiety, and it fails to encode multiple behavioral contexts that do not. Strikingly, the activity of this network is also principally altered in two mouse models of depression. Thus, we establish a network-level process whereby the brain encodes anxiety in health and disease.
Collapse
Affiliation(s)
- Dalton N Hughes
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Michael Hunter Klein
- Dept. of Electrical and Computer Engineering, Duke University, Durham North Carolina 27708, USA
| | | | - Gwenaëlle E Thomas
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yael Grossman
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Diana Waters
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Anna E Matthews
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - William E Carson
- Dept. of Biomedical Engineering, Duke University, Durham North Carolina 27708, USA
| | - Yassine Filali
- Department of Molecular Physiology and Biophysics, Department of Psychiatry, University of Iowa, Iowa City, IA, 52242 USA
| | - Mariya Tsyglakova
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA 15213
| | - Alexandra Fink
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Neil M Gallagher
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Masiel Perez-Balaguer
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA 15213
| | - Jean Mary Zarate
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Rainbo C Hultman
- Department of Molecular Physiology and Biophysics, Department of Psychiatry, University of Iowa, Iowa City, IA, 52242 USA
| | - Stephen D Mague
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David E Carlson
- Dept. of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Electrical and Computer Engineering, Duke University, Durham North Carolina 27708, USA
- Dept. of Civil and Environmental Engineering, Duke University, Durham North Carolina 27708, USA
- Dept. of Biomedical Engineering, Duke University, Durham North Carolina 27708, USA
| | - Kafui Dzirasa
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Neurosurgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
3
|
Kong Q, Li F, Sun K, Sun X, Ma J. Valproic acid ameliorates cauda equina injury by suppressing HDAC2-mediated ferroptosis. CNS Neurosci Ther 2024; 30:e14524. [PMID: 38105511 PMCID: PMC11017456 DOI: 10.1111/cns.14524] [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: 08/18/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 12/19/2023] Open
Abstract
INTRODUCTION Persistent neuroinflammatory response after cauda equina injury (CEI) lowers nociceptor firing thresholds, accompanied by pathological pain and decreasing extremity dysfunction. Histone deacetylation has been considered a key regulator of immunity, inflammation, and neurological dysfunction. Our previous study suggested that valproic acid (VPA), a histone deacetylase inhibitor, exhibited neuroprotective effects in rat models of CEI, although the underlying mechanism remains elusive. METHODS The cauda equina compression surgery was performed to establish the CEI model. The Basso, Beattie, Bresnahan score, and the von Frey filament test were carried out to measure the animal behavior. Immunofluorescence staining of myelin basic protein and GPX4 was carried out. In addition, transmission electron microscope analysis was used to assess the effect of VPA on the morphological changes of mitochondria. RNA-sequencing was conducted to clarify the underlying mechanism of VPA on CEI protection. RESULTS In this current study, we revealed that the expression level of HDAC1 and HDAC2 was elevated after cauda equina compression model but was reversed by VPA treatment. Meanwhile, HDAC2 knockdown resulted in the improvement of motor functions and pathologic pain, similar to treatment with VPA. Histology analysis also showed that knockdown of histone deacetylase (HDAC)-2, but not HDAC1, remarkably alleviated cauda equina injury and demyelinating lesions. The potential mechanism may be related to lowering oxidative stress and inflammatory response in the injured region. Notably, the transcriptome sequencing indicated that the therapeutic effect of VPA may depend on HDAC2-mediated ferroptosis. Ferroptosis-related genes were analyzed in vivo and DRG cells further validated the reliability of RNA-sequencing results, suggesting HDAC2-H4K12ac axis participated in epigenetic modulation of ferroptosis-related genes. CONCLUSION HDAC2 is critically involved in the ferroptosis and neuroinflammation in cauda equina injury, and VPA ameliorated cauda equina injury by suppressing HDAC2-mediated ferroptosis.
Collapse
Affiliation(s)
- Qingjie Kong
- Department of OrthopedicsShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Key Laboratory of Medical Immunology & Institute of ImmunologySecond Military Medical UniversityShanghaiChina
| | - Fudong Li
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Kaiqiang Sun
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Xiaofei Sun
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Jun Ma
- Department of OrthopedicsShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
4
|
Dollish HK, Tsyglakova M, McClung CA. Circadian rhythms and mood disorders: Time to see the light. Neuron 2024; 112:25-40. [PMID: 37858331 PMCID: PMC10842077 DOI: 10.1016/j.neuron.2023.09.023] [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: 06/01/2023] [Revised: 08/09/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
The importance of time is ever prevalent in our world, and disruptions to the normal light/dark and sleep/wake cycle have now become the norm rather than the exception for a large part of it. All mood disorders, including seasonal affective disorder (SAD), major depressive disorder (MDD), and bipolar disorder (BD), are strongly associated with abnormal sleep and circadian rhythms in a variety of physiological processes. Environmental disruptions to normal sleep/wake patterns, light/dark changes, and seasonal changes can precipitate episodes. Moreover, treatments that target the circadian system have proven to be therapeutic in certain cases. This review will summarize much of our current knowledge of how these disorders associate with specific circadian phenotypes, as well as the neuronal mechanisms that link the circadian clock with mood regulation. We also discuss what has been learned from therapies that target circadian rhythms and how we may use current knowledge to develop more individually designed treatments.
Collapse
Affiliation(s)
- Hannah K Dollish
- Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, USA
| | - Mariya Tsyglakova
- Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, USA
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, USA.
| |
Collapse
|
5
|
Bhatnagar A, Murray G, Ray S. Circadian biology to advance therapeutics for mood disorders. Trends Pharmacol Sci 2023; 44:689-704. [PMID: 37648611 DOI: 10.1016/j.tips.2023.07.008] [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: 05/21/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 09/01/2023]
Abstract
Mood disorders account for a significant global disease burden, and pharmacological innovation is needed as existing medications are suboptimal. A wide range of evidence implicates circadian and sleep dysfunction in the pathogenesis of mood disorders, and there is growing interest in these chronobiological pathways as a focus for treatment innovation. We review contemporary evidence in three promising areas in circadian-clock-based therapeutics in mood disorders: targeting the circadian system informed by mechanistic molecular advances; time-tailoring of medications; and personalizing treatment using circadian parameters. We also consider the limitations and challenges in accelerating the development of new circadian-informed pharmacotherapies for mood disorders.
Collapse
Affiliation(s)
- Apoorva Bhatnagar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India; Centre for Mental Health, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Greg Murray
- Centre for Mental Health, Swinburne University of Technology, Melbourne, Victoria, Australia.
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India.
| |
Collapse
|
6
|
Wen J, Chen Y, Yang J, Dai C, Yu S, Zhong W, Liu L, He C, Zhang W, Yang T, Liu L, Hu J. Valproic acid increases CAR T cell cytotoxicity against acute myeloid leukemia. J Immunother Cancer 2023; 11:e006857. [PMID: 37524506 PMCID: PMC10391797 DOI: 10.1136/jitc-2023-006857] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/02/2023] Open
Abstract
The treatment of B cell malignancies has dramatically changed with the introduction of immunotherapy, especially chimeric antigen receptor T (CAR-T) cell therapy. However, only limited efficacy is observed in acute myeloid leukaemia (AML). In the study, We detected CD123 and CLL-1 expression on leukaemia cells from Relapsed/Refractory AML (R/R AML) patients. Then, we constructed anti-CD123 CAR and CLL-1 CAR with different co-stimulation domains (CD28 or 4-1BB) and detected their anti-AML effects. To increase the efficacy of CAR-T cell therapy, we tested different strategies, including application of combined checkpoint inhibitors and histone deacetylase inhibitors (HDACi) in vivo and in vitro We found CD123 and CLL-1 were highly expressed on AML cells. The proportions of T cell subsets and NK cells involved in anti-tumour or anti-inflammation processes in AML patients significantly decreased when compared with healthy donors. Both CD123 CAR and CLL-1 CAR displayed specific anti-AML effects in vitro To improve the lysis effects of CAR-T cells, we combined CAR-T cell therapy with different agents. PD-1/PD-L1 antibodies only slightly improved the potency of CAR-T cell therapy (CD123 CAR-T 60.92% ± 2.9087% vs. 65.43% ± 2.1893%, 60.92% ± 2.9087% vs. 67.43% ± 3.4973%; 37.37% ± 3.908% vs. 41.89% ± 5.1568%, 37.37% ± 3.908% vs. 42.84% ± 4.2635%). However, one HDACi (valproic acid [VPA]) significantly improved CAR-T cell potency against AML cells (CLL-1 CAR-T 34.97% ± 0.3051% vs. 88.167% ± 1.5327%, p < 0.0001; CD123 CAR-T 26.87% ± 2.7010% vs. 82.56% ± 3.086%, p < 0.0001 in MV411; CLL-1 CAR-T 78.77% ± 1.2061% vs. 93.743% ± 1.2333%, p < 0.0001; CD123 CAR-T 64.10% ± 1.5130% vs. 94.427% ± 0.142%, p = 0.0001 in THP-1). Combination therapy prolonged the overall survival of mice when compared with single CD123 CAR-T cell therapy (median survival: 180 days vs. unfollowed). A possible mechanism is that activated CD8+T cells upregulate natural-killer group 2 member D (NKG2D), and VPA upregulates NKG2D ligand expression in AML cells, contributing to NKG2D-mediated cytotoxicity of CAR-T cells against tumour cells. In conclusion, CD123 and CLL-1 are promising targets for AML CAR-T cell therapy. A combination of VPA pre-treatment and CAR-T against AML exhibits synergic effects.
Collapse
MESH Headings
- Animals
- Mice
- Valproic Acid/pharmacology
- Valproic Acid/therapeutic use
- Receptors, Chimeric Antigen/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- T-Lymphocytes
Collapse
Affiliation(s)
- Jingjing Wen
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Department of Lymphoma, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| | - Yanxin Chen
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Jiajie Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chunye Dai
- School of Life Sciences, Fudan University, Shanghai, China
| | - Shenjie Yu
- Department of Internal medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenting Zhong
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Lilin Liu
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Chengguanng He
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Wenmin Zhang
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
- Pathological Diagnosis Center & Oncology Institution, Fujian Medical University, Fuzhou, China
| | - Ting Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Lingfeng Liu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Jianda Hu
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
7
|
Peedicayil J. Genome-Environment Interactions and Psychiatric Disorders. Biomedicines 2023; 11:biomedicines11041209. [PMID: 37189827 DOI: 10.3390/biomedicines11041209] [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: 03/27/2023] [Revised: 04/08/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Environmental factors are known to interact with the genome by altering epigenetic mechanisms regulating gene expression and contributing to the pathogenesis of psychiatric disorders. This article is a narrative review of how the major environmental factors contribute to the pathogenesis of common psychiatric disorders such as schizophrenia, bipolar disorder, major depressive disorder, and anxiety disorder this way. The cited articles were published between 1 January 2000 and 31 December 2022 and were obtained from PubMed and Google Scholar. The search terms used were as follows: gene or genetic; genome; environment; mental or psychiatric disorder; epigenetic; and interaction. The following environmental factors were found to act epigenetically on the genome to influence the pathogenesis of psychiatric disorders: social determinants of mental health, maternal prenatal psychological stress, poverty, migration, urban dwelling, pregnancy and birth complications, alcohol and substance abuse, microbiota, and prenatal and postnatal infections. The article also discusses the ways by which factors such as drugs, psychotherapy, electroconvulsive therapy, and physical exercise act epigenetically to alleviate the symptoms of psychiatric disorders in affected patients. These data will be useful information for clinical psychiatrists and those researching the pathogenesis and treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Jacob Peedicayil
- Department of Pharmacology & Clinical Pharmacology, Christian Medical College, Vellore 632 002, India
| |
Collapse
|
8
|
Albrecht U. The circadian system and mood related behavior in mice. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:269-291. [PMID: 37709379 DOI: 10.1016/bs.apcsb.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Most organisms on earth have evolved an internal clock in order to predict daily recurring events. This clock called circadian clock has a period of about 24 h and allows organisms to organize biochemical and physiological processes over one day. Changes in lighting conditions as they occur naturally over seasons, or man made by jet lag or shift work, advance or delay clock phase in order to synchronize an organism's physiology to the environment. A misalignment of the clock to its environment results in sleep disturbances and mood disorders. Although there are strong associations between the circadian clock and mood disorders such as depression, the underlying molecular mechanisms are not well understood. This review describes the currently known molecular links between circadian clock components and mood related behaviors in mice, which will help to understand the causal links between the clock and mood in humans in the future.
Collapse
Affiliation(s)
- U Albrecht
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
9
|
Kyzar EJ, Denfield GH. Taking subjectivity seriously: towards a unification of phenomenology, psychiatry, and neuroscience. Mol Psychiatry 2023; 28:10-16. [PMID: 36460728 PMCID: PMC10130907 DOI: 10.1038/s41380-022-01891-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022]
Abstract
Nearly all psychiatric diseases involve alterations in subjective, lived experience. The scientific study of the biological basis of mental illness has generally focused on objective measures and observable behaviors, limiting the potential for our understanding of brain mechanisms of disease states and possible treatments. However, applying methods designed principally to interpret objective behavioral measures to the measurement and extrapolation of subjective states presents a number of challenges. In order to help bridge this gap, we draw on the tradition of phenomenology, a philosophical movement concerned with elucidating the structure of lived experience, which emerged in the early 20th century and influenced philosophy of mind, cognitive science, and psychiatry. A number of early phenomenologically-oriented psychiatrists made influential contributions to the field, but this approach retreated to the background as psychiatry moved towards more operationalized disease classifications. Recently, clinical-phenomenological research and viewpoints have re-emerged in the field. We argue that the potential for phenomenological research and methods to generate productive hypotheses about the neurobiological basis of psychiatric diseases has thus far been underappreciated. Using specific examples drawing on the subjective experience of mania and psychosis, we demonstrate that phenomenologically-oriented clinical studies can generate novel and fruitful propositions for neuroscientific investigation. Additionally, we outline a proposal for more rigorously integrating phenomenological investigations of subjective experience with the methods of modern neuroscience research, advocating a cross-species approach with a key role for human subjects research. Collaborative interaction between phenomenology, psychiatry, and neuroscience has the potential to move these fields towards a unified understanding of the biological basis of mental illness.
Collapse
Affiliation(s)
- Evan J Kyzar
- Department of Psychiatry, Columbia University, New York, NY, USA. .,Research Foundation for Mental Hygiene, Menands, NY, USA. .,New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, USA.
| | - George H Denfield
- Department of Psychiatry, Columbia University, New York, NY, USA. .,Research Foundation for Mental Hygiene, Menands, NY, USA. .,New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, USA.
| |
Collapse
|
10
|
Micale V, Di Bartolomeo M, Di Martino S, Stark T, Dell'Osso B, Drago F, D'Addario C. Are the epigenetic changes predictive of therapeutic efficacy for psychiatric disorders? A translational approach towards novel drug targets. Pharmacol Ther 2023; 241:108279. [PMID: 36103902 DOI: 10.1016/j.pharmthera.2022.108279] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
The etiopathogenesis of mental disorders is not fully understood and accumulating evidence support that clinical symptomatology cannot be assigned to a single gene mutation, but it involves several genetic factors. More specifically, a tight association between genes and environmental risk factors, which could be mediated by epigenetic mechanisms, may play a role in the development of mental disorders. Several data suggest that epigenetic modifications such as DNA methylation, post-translational histone modification and interference of microRNA (miRNA) or long non-coding RNA (lncRNA) may modify the severity of the disease and the outcome of the therapy. Indeed, the study of these mechanisms may help to identify patients particularly vulnerable to mental disorders and may have potential utility as biomarkers to facilitate diagnosis and treatment of psychiatric disorders. This article summarizes the most relevant preclinical and human data showing how epigenetic modifications can be central to the therapeutic efficacy of antidepressant and/or antipsychotic agents, as possible predictor of drugs response.
Collapse
Affiliation(s)
- Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Martina Di Bartolomeo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Serena Di Martino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Tibor Stark
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Scientific Core Unit Neuroimaging, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bernardo Dell'Osso
- Department of Biomedical and Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy, Department of Mental Health, ASST Fatebenefratelli-Sacco, Milan, Italy; "Aldo Ravelli" Research Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan Medical School, Milan, Italy; Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Guo L, Qi YJ, Tan H, Dai D, Balesar R, Sluiter A, van Heerikhuize J, Hu SH, Swaab DF, Bao AM. Different oxytocin and corticotropin-releasing hormone system changes in bipolar disorder and major depressive disorder patients. EBioMedicine 2022; 84:104266. [PMID: 36126617 PMCID: PMC9489957 DOI: 10.1016/j.ebiom.2022.104266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/11/2022] Open
Abstract
Background Oxytocin (OXT) and corticotropin-releasing hormone (CRH) are both produced in hypothalamic paraventricular nucleus (PVN). Central CRH may cause depression-like symptoms, while peripheral higher OXT plasma levels were proposed to be a trait marker for bipolar disorder (BD). We aimed to investigate differential OXT and CRH expression in the PVN and their receptors in prefrontal cortex of major depressive disorder (MDD) and BD patients. In addition, we investigated mood-related changes by stimulating PVN-OXT in mice. Methods Quantitative immunocytochemistry and in situ hybridization were performed in the PVN for OXT and CRH on 6 BD and 6 BD-controls, 9 MDD and 9 MDD-controls. mRNA expressions of their receptors (OXTR, CRHR1 and CRHR2) were determined in anterior cingulate cortex and dorsolateral prefrontal cortex (DLPFC) of 30 BD and 34 BD-controls, and 24 MDD and 12 MDD-controls. PVN of 41 OXT-cre mice was short- or long-term activated by chemogenetics, and mood-related behavior was compared with 26 controls. Findings Significantly increased OXT-immunoreactivity (ir), OXT-mRNA in PVN and increased OXTR-mRNA in DLPFC, together with increased ratios of OXT-ir/CRH-ir and OXTR-mRNA/CRHR-mRNA were observed in BD, at least in male BD patients, but not in MDD patients. PVN-OXT stimulation induced depression-like behaviors in male mice, and mixed depression/mania-like behaviors in female mice in a time-dependent way. Interpretation Increased PVN-OXT and DLPFC-OXTR expression are characteristic for BD, at least for male BD patients. Stimulation of PVN-OXT neurons induced mood changes in mice, in a pattern different from BD. Funding 10.13039/501100001809National Natural Science Foundation of China (81971268, 82101592).
Collapse
|
12
|
Singla R, Mishra A, Cao R. The trilateral interactions between mammalian target of rapamycin (mTOR) signaling, the circadian clock, and psychiatric disorders: an emerging model. Transl Psychiatry 2022; 12:355. [PMID: 36045116 PMCID: PMC9433414 DOI: 10.1038/s41398-022-02120-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Circadian (~24 h) rhythms in physiology and behavior are evolutionarily conserved and found in almost all living organisms. The rhythms are endogenously driven by daily oscillatory activities of so-called "clock genes/proteins", which are widely distributed throughout the mammalian brain. Mammalian (mechanistic) target of rapamycin (mTOR) signaling is a fundamental intracellular signal transduction cascade that controls important neuronal processes including neurodevelopment, synaptic plasticity, metabolism, and aging. Dysregulation of the mTOR pathway is associated with psychiatric disorders including autism spectrum disorders (ASD) and mood disorders (MD), in which patients often exhibit disrupted daily physiological rhythms and abnormal circadian gene expression in the brain. Recent work has found that the activities of mTOR signaling are temporally controlled by the circadian clock and exhibit robust circadian oscillations in multiple systems. In the meantime, mTOR signaling regulates fundamental properties of the central and peripheral circadian clocks, including period length, entrainment, and synchronization. Whereas the underlying mechanisms remain to be fully elucidated, increasing clinical and preclinical evidence support significant crosstalk between mTOR signaling, the circadian clock, and psychiatric disorders. Here, we review recent progress in understanding the trilateral interactions and propose an "interaction triangle" model between mTOR signaling, the circadian clock, and psychiatric disorders (focusing on ASD and MD).
Collapse
Affiliation(s)
- Rubal Singla
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Abhishek Mishra
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA. .,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
13
|
Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry 2022; 27:2927-2939. [PMID: 35351989 DOI: 10.1038/s41380-022-01523-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/25/2022]
Abstract
Genome-wide association studies (GWAS) have reported substantial genomic loci significantly associated with clinical risk of bipolar disorder (BD), and studies combining techniques of genetics, neuroscience, neuroimaging, and pharmacology are believed to help tackle clinical problems (e.g., identifying novel therapeutic targets). However, translating findings of psychiatric genetics into biological mechanisms underlying BD pathogenesis remains less successful. Biological impacts of majority of BD GWAS risk loci are obscure, and the involvement of many GWAS risk genes in this illness is yet to be investigated. It is thus necessary to review the progress of applying BD GWAS risk genes in the research and intervention of the disorder. A comprehensive literature search found that a number of such risk genes had been investigated in cellular or animal models, even before they were highlighted in BD GWAS. Intriguingly, manipulation of many BD risk genes (e.g., ANK3, CACNA1C, CACNA1B, HOMER1, KCNB1, MCHR1, NCAN, SHANK2 etc.) resulted in altered murine behaviors largely restoring BD clinical manifestations, including mania-like symptoms such as hyperactivity, anxiolytic-like behavior, as well as antidepressant-like behavior, and these abnormalities could be attenuated by mood stabilizers. In addition to recapitulating phenotypic characteristics of BD, some GWAS risk genes further provided clues for the neurobiology of this illness, such as aberrant activation and functional connectivity of brain areas in the limbic system, and modulated dendritic spine morphogenesis as well as synaptic plasticity and transmission. Therefore, BD GWAS risk genes are undoubtedly pivotal resources for modeling this illness, and might be translational therapeutic targets in the future clinical management of BD. We discuss both promising prospects and cautions in utilizing the bulk of useful resources generated by GWAS studies. Systematic integrations of findings from genetic and neuroscience studies are called for to promote our understanding and intervention of BD.
Collapse
|
14
|
McCarthy MJ, Gottlieb JF, Gonzalez R, McClung CA, Alloy LB, Cain S, Dulcis D, Etain B, Frey BN, Garbazza C, Ketchesin KD, Landgraf D, Lee H, Marie‐Claire C, Nusslock R, Porcu A, Porter R, Ritter P, Scott J, Smith D, Swartz HA, Murray G. Neurobiological and behavioral mechanisms of circadian rhythm disruption in bipolar disorder: A critical multi-disciplinary literature review and agenda for future research from the ISBD task force on chronobiology. Bipolar Disord 2022; 24:232-263. [PMID: 34850507 PMCID: PMC9149148 DOI: 10.1111/bdi.13165] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM Symptoms of bipolar disorder (BD) include changes in mood, activity, energy, sleep, and appetite. Since many of these processes are regulated by circadian function, circadian rhythm disturbance has been examined as a biological feature underlying BD. The International Society for Bipolar Disorders Chronobiology Task Force (CTF) was commissioned to review evidence for neurobiological and behavioral mechanisms pertinent to BD. METHOD Drawing upon expertise in animal models, biomarkers, physiology, and behavior, CTF analyzed the relevant cross-disciplinary literature to precisely frame the discussion around circadian rhythm disruption in BD, highlight key findings, and for the first time integrate findings across levels of analysis to develop an internally consistent, coherent theoretical framework. RESULTS Evidence from multiple sources implicates the circadian system in mood regulation, with corresponding associations with BD diagnoses and mood-related traits reported across genetic, cellular, physiological, and behavioral domains. However, circadian disruption does not appear to be specific to BD and is present across a variety of high-risk, prodromal, and syndromic psychiatric disorders. Substantial variability and ambiguity among the definitions, concepts and assumptions underlying the research have limited replication and the emergence of consensus findings. CONCLUSIONS Future research in circadian rhythms and its role in BD is warranted. Well-powered studies that carefully define associations between BD-related and chronobiologically-related constructs, and integrate across levels of analysis will be most illuminating.
Collapse
Affiliation(s)
- Michael J. McCarthy
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
- VA San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - John F. Gottlieb
- Department of PsychiatryFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Robert Gonzalez
- Department of Psychiatry and Behavioral HealthPennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Colleen A. McClung
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lauren B. Alloy
- Department of PsychologyTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sean Cain
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityMelbourneVictoriaAustralia
| | - Davide Dulcis
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | - Bruno Etain
- Université de ParisINSERM UMR‐S 1144ParisFrance
| | - Benicio N. Frey
- Department Psychiatry and Behavioral NeuroscienceMcMaster UniversityHamiltonOntarioCanada
| | - Corrado Garbazza
- Centre for ChronobiologyPsychiatric Hospital of the University of Basel and Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
| | - Kyle D. Ketchesin
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dominic Landgraf
- Circadian Biology GroupDepartment of Molecular NeurobiologyClinic of Psychiatry and PsychotherapyUniversity HospitalLudwig Maximilian UniversityMunichGermany
| | - Heon‐Jeong Lee
- Department of Psychiatry and Chronobiology InstituteKorea UniversitySeoulSouth Korea
| | | | - Robin Nusslock
- Department of Psychology and Institute for Policy ResearchNorthwestern UniversityChicagoIllinoisUSA
| | - Alessandra Porcu
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | | | - Philipp Ritter
- Clinic for Psychiatry and PsychotherapyCarl Gustav Carus University Hospital and Technical University of DresdenDresdenGermany
| | - Jan Scott
- Institute of NeuroscienceNewcastle UniversityNewcastleUK
| | - Daniel Smith
- Division of PsychiatryUniversity of EdinburghEdinburghUK
| | - Holly A. Swartz
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Greg Murray
- Centre for Mental HealthSwinburne University of TechnologyMelbourneVictoriaAustralia
| |
Collapse
|
15
|
Seney ML, Kim SM, Glausier JR, Hildebrand MA, Xue X, Zong W, Wang J, Shelton MA, Phan BN, Srinivasan C, Pfenning AR, Tseng GC, Lewis DA, Freyberg Z, Logan RW. Transcriptional Alterations in Dorsolateral Prefrontal Cortex and Nucleus Accumbens Implicate Neuroinflammation and Synaptic Remodeling in Opioid Use Disorder. Biol Psychiatry 2021; 90:550-562. [PMID: 34380600 PMCID: PMC8463497 DOI: 10.1016/j.biopsych.2021.06.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prevalence rates of opioid use disorder (OUD) have increased dramatically, accompanied by a surge of overdose deaths. While opioid dependence has been extensively studied in preclinical models, an understanding of the biological alterations that occur in the brains of people who chronically use opioids and who are diagnosed with OUD remains limited. To address this limitation, RNA sequencing was conducted on the dorsolateral prefrontal cortex and nucleus accumbens, regions heavily implicated in OUD, from postmortem brains in subjects with OUD. METHODS We performed RNA sequencing on the dorsolateral prefrontal cortex and nucleus accumbens from unaffected comparison subjects (n = 20) and subjects diagnosed with OUD (n = 20). Our transcriptomic analyses identified differentially expressed transcripts and investigated the transcriptional coherence between brain regions using rank-rank hypergeometric orderlap. Weighted gene coexpression analyses identified OUD-specific modules and gene networks. Integrative analyses between differentially expressed transcripts and genome-wide association study datasets using linkage disequilibrium scores assessed the genetic liability of psychiatric-related phenotypes in OUD. RESULTS Rank-rank hypergeometric overlap analyses revealed extensive overlap in transcripts between the dorsolateral prefrontal cortex and nucleus accumbens in OUD, related to synaptic remodeling and neuroinflammation. Identified transcripts were enriched for factors that control proinflammatory cytokine, chondroitin sulfate, and extracellular matrix signaling. Cell-type deconvolution implicated a role for microglia as a potential driver for opioid-induced neuroplasticity. Linkage disequilibrium score analysis suggested genetic liabilities for risky behavior, attention-deficit/hyperactivity disorder, and depression in subjects with OUD. CONCLUSIONS Overall, our findings suggest connections between the brain's immune system and opioid dependence in the human brain.
Collapse
Affiliation(s)
- Marianne L Seney
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sam-Moon Kim
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine
| | - Jill R Glausier
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jiebiao Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Micah A Shelton
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - BaDoi N Phan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Chaitanya Srinivasan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Andreas R Pfenning
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan W Logan
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts; Center for Systems Neuroscience, Boston University, Boston, Massachusetts.
| |
Collapse
|
16
|
Enlightened: addressing circadian and seasonal changes in photoperiod in animal models of bipolar disorder. Transl Psychiatry 2021; 11:373. [PMID: 34226504 PMCID: PMC8257630 DOI: 10.1038/s41398-021-01494-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
Bipolar disorders (BDs) exhibit high heritability and symptoms typically first occur during late adolescence or early adulthood. Affected individuals may experience alternating bouts of mania/hypomania and depression, with euthymic periods of varying lengths interspersed between these extremes of mood. Clinical research studies have consistently demonstrated that BD patients have disturbances in circadian and seasonal rhythms, even when they are free of symptoms. In addition, some BD patients display seasonal patterns in the occurrence of manic/hypomanic and depressive episodes as well as the time of year when symptoms initially occur. Finally, the age of onset of BD symptoms is strongly influenced by the distance one lives from the equator. With few exceptions, animal models useful in the study of BD have not capitalized on these clinical findings regarding seasonal patterns in BD to explore molecular mechanisms associated with the expression of mania- and depression-like behaviors in laboratory animals. In particular, animal models would be especially useful in studying how rates of change in photoperiod that occur during early spring and fall interact with risk genes to increase the occurrence of mania- and depression-like phenotypes, respectively. Another unanswered question relates to the ways in which seasonally relevant changes in photoperiod affect responses to acute and chronic stressors in animal models. Going forward, we suggest ways in which translational research with animal models of BD could be strengthened through carefully controlled manipulations of photoperiod to enhance our understanding of mechanisms underlying seasonal patterns of BD symptoms in humans. In addition, we emphasize the value of incorporating diurnal rodent species as more appropriate animal models to study the effects of seasonal changes in light on symptoms of depression and mania that are characteristic of BD in humans.
Collapse
|