1
|
Tao Y, Pan Q, Cai T, Lu ZH, Haque M, Dottorini T, Colvin LA, Smith BH, Meng W. A genome-wide association study identifies novel genetic variants associated with neck or shoulder pain in the UK biobank (N = 430,193). Pain Rep 2025; 10:e1267. [PMID: 40291381 PMCID: PMC12026381 DOI: 10.1097/pr9.0000000000001267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/09/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Neck and shoulder pain are prevalent musculoskeletal disorders that significantly affect the quality of life for a substantial portion of the global population. Studies have shown that women are more susceptible than men. Objective This study aims to discover genetic variants associated with neck or shoulder pain through a genome-wide association study (GWAS), using data from 430,193 participants in the UK Biobank. Methods A genome-wide association study was performed adjusting for age, sex, BMI, and 8 population principal components. Significant and independent genetic variants were replicated by FinnGen. Results The primary GWAS revealed 5 significant genetic loci (including 2 novel) associated with neck or shoulder pain, with the most significant single nucleotide polymorphism (SNP) being rs9889282 (P = 2.63 × 10-12) near CA10 on chromosome 17. Two novel significant associations were detected on chromosomes 18 and 14, with the top SNPs being rs4608411 (P = 8.20 × 10-9) near TCF4 and rs370565192 (P = 3.80 × 10-8) in DCAF5, respectively. Our secondary GWAS identified a single novel genetic locus in SLC24A3 among males and 2 genetic loci (including one novel near LINC02770) among females. In the replication stage, the SLC39A8 locus was weakly supported by the FinnGen cohort. The tissue expression analysis revealed a significant association between brain tissues and neck or shoulder pain. Conclusion In summary, this study has identified novel genetic variants for neck or shoulder pain. Sex-stratified GWAS also suggested that sex played a role in the occurrence of the phenotype.
Collapse
Affiliation(s)
- Yiwen Tao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Qi Pan
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Zen Huat Lu
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei Darussalam
| | - Mainul Haque
- School of Mathematical Sciences, University of Nottingham Ningbo China, Ningbo, China
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Lesley A. Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Blair H. Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
- Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
2
|
Shil A, Arava N, Levi N, Levine L, Golan H, Meiri G, Michaelovski A, Tsadaka Y, Aran A, Menashe I. An integrative scoring approach for prioritization of rare autism spectrum disorder candidate variants from whole exome sequencing data. Sci Rep 2025; 15:13024. [PMID: 40234546 PMCID: PMC12000308 DOI: 10.1038/s41598-025-96063-x] [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: 06/04/2024] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
Discerning clinically relevant autism spectrum disorder (ASD) candidate variants from whole-exome sequencing (WES) data is complex, time-consuming, and labor-intensive. To this end, we developed AutScore, an integrative prioritization algorithm of ASD candidate variants from WES data and assessed its performance to detect clinically relevant variants. We studied WES data from 581 ASD probands, and their parents registered in the Azrieli National Center database for Autism and Neurodevelopment Research. We focused on rare allele frequency (< 1%) and high-quality proband-specific variants affecting genes associated with ASD or other neurodevelopmental disorders (NDDs). We developed AutScore and AutScore.r and assigned each variant based on their pathogenicity, clinical relevance, gene-disease association, and inheritance patterns. Finally, we compared the performance of both AutScore versions with the rating of clinical experts and the NDD variant prioritization algorithm, AutoCaSc. Overall, 1161 rare variants distributed in 687 genes in 441 ASD probands were evaluated by AutScore with scores ranging from - 4 to 25, with a mean ± SD of 5.89 ± 4.18. AutScore.r cut-off of ≥ 0.335 performs better than AutoCaSc and AutScore in detecting clinically relevant ASD variants, with a detection accuracy rate of 85% and an overall diagnostic yield of 10.3%. Five variants with AutScore.r of ≥ 0.335 were distributed in five novel ASD candidate genes. AutScore.r is an effective automated ranking system for ASD candidate variants that could be implemented in ASD clinical genetics pipelines.
Collapse
Affiliation(s)
- Apurba Shil
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Noa Arava
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Noam Levi
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Liron Levine
- Bioinformatics Core Facility, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hava Golan
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Gal Meiri
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Preschool Psychiatric Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Analya Michaelovski
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Child Development Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Yair Tsadaka
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Child Development Center, Ministry of Health, Be'er Sheva, 84100, Israel
| | - Adi Aran
- Neuropediatric Unit, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idan Menashe
- Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Azrieli National Centre for Autism and Neurodevelopment Research, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
3
|
Uçak EF, Altınbaş K, Koçak N, Güleç A. Circadian rhythm and lithium response in bipolar disorder: Insights from actigraphy and NR1D1 polymorphism. Chronobiol Int 2025; 42:225-234. [PMID: 39831746 DOI: 10.1080/07420528.2025.2455139] [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: 08/08/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Lithium has long been used as a cornerstone mood stabilizer in the treatment of bipolar disorder (BD). However, reliable biomarkers that can predict which patients will respond better to lithium are still lacking. This study aims to evaluate the potential of NR1D1 gene SNP; rs2071427 and actigraphic measurements in predicting lithium response. Thirty-one patients diagnosed with BD at Selçuk University Faculty of Medicine and who were euthymic for at least 8 weeks were included in the study. Sleep-wake cycles and circadian rhythms of the participants were monitored by actigraph for approximately 1 week. For genetic analyses, the SNP rs2071427 variant of the NR1D1 gene was evaluated. A significant proportion of patients with homozygous (AA/GG) genotypes responded well to lithium, whereas some patients with heterozygous (AG) genotypes did not respond to lithium. Actigraphic data showed that there were marked variations in the sleep patterns of BD patients. The Morningness-Eveningness Questionnaire scale did not adequately discriminate the morning chronotype. Seasonal Pattern Assessment Questionnaire results showed that most patients had a seasonal pattern, but this was insufficient to predict response to lithium. This study once again demonstrates the need for new biomarkers to predict lithium response. The findings are an important step in the personalization of BD treatment and may improve treatment efficacy and minimize side effects by tailoring the treatment process to the individual characteristics of patients. Future studies should support these findings with larger sample groups and studies on different genetic markers.
Collapse
Affiliation(s)
- Ekrem Furkan Uçak
- Department of Psychiatry, Afyonkarahisar Health Sciences University, Zafer Sağlık Külliyesi Dörtyol Mah, Afyonkarahisar, Turkey
| | - Kürşat Altınbaş
- Department of Psychiatry, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Nadir Koçak
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ahmet Güleç
- Department of Child and Adolescent Psychiatry, Balıkesir Atatürk City Hospital, Balıkesir, Turkey
| |
Collapse
|
4
|
McGrouther CC, Rangan AV, Di Florio A, Elman JA, Schork NJ, Kelsoe J, Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. Heterogeneity analysis provides evidence for a genetically homogeneous subtype of bipolar-disorder. PLoS One 2025; 20:e0314288. [PMID: 39879180 PMCID: PMC11778664 DOI: 10.1371/journal.pone.0314288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/07/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Bipolar Disorder (BD) is a complex disease. It is heterogeneous, both at the phenotypic and genetic level, although the extent and impact of this heterogeneity is not fully understood. One way to assess this heterogeneity is to look for patterns in the subphenotype data. Because of the variability in how phenotypic data was collected by the various BD studies over the years, homogenizing this subphenotypic data is a challenging task, and so is replication. An alternative methodology, taken here, is to set aside the intricacies of subphenotype and allow the genetic data itself to determine which subjects define a homogeneous genetic subgroup (termed 'bicluster' below). RESULTS In this paper, we leverage recent advances in heterogeneity analysis to look for genetically-driven subgroups (i.e., biclusters) within the broad phenotype of Bipolar Disorder. We first apply this covariate-corrected biclustering algorithm to a cohort of 2524 BD cases and 4106 controls from the Bipolar Disease Research Network (BDRN) within the Psychiatric Genomics Consortium (PGC). We find evidence of genetic heterogeneity delineating a statistically significant bicluster comprising a subset of BD cases which exhibits a disease-specific pattern of differential-expression across a subset of SNPs. This disease-specific genetic pattern (i.e., 'genetic subgroup') replicates across the remaining data-sets collected by the PGC containing 5781/8289, 3581/7591, and 6825/9752 cases/controls, respectively. This genetic subgroup (discovered without using any BD subtype information) was more prevalent in Bipolar type-I than in Bipolar type-II. CONCLUSIONS Our methodology has successfully identified a replicable homogeneous genetic subgroup of bipolar disorder. This subgroup may represent a collection of correlated genetic risk-factors for BDI. By investigating the subgroup's bicluster-informed polygenic-risk-scoring (PRS), we find that the disease-specific pattern highlighted by the bicluster can be leveraged to eliminate noise from our GWAS analyses and improve risk prediction. This improvement is particularly notable when using only a relatively small subset of the available SNPs, implying improved SNP replication. Though our primary focus is only the analysis of disease-related signal, we also identify replicable control-related heterogeneity.
Collapse
Affiliation(s)
- Caroline C. McGrouther
- Courant Institute of Mathematical Sciences, New York University, New York, NY, United States of America
| | - Aaditya V. Rangan
- Courant Institute of Mathematical Sciences, New York University, New York, NY, United States of America
| | - Arianna Di Florio
- School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Jeremy A. Elman
- Department of Psychiatry, University of California San Diego, San Diego, CA, United States of America
| | - Nicholas J. Schork
- The Translational Genomics Research Institute, Quantitative Medicine and Systems Biology, Phoenix, AZ, United States of America
| | - John Kelsoe
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America
| | | |
Collapse
|
5
|
Çolak-Geniş E, Özdemir Erdoğan M, Çam FS, Aydemir Ö, Akin F, Gerik-Celebi HB, Solak M. Investigation of Genetic Changes in Three Families with Bipolar Disease. Mol Syndromol 2024; 15:464-473. [PMID: 39634238 PMCID: PMC11614438 DOI: 10.1159/000539115] [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: 12/25/2023] [Accepted: 04/25/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Bipolar disorder (BD) is a serious psychiatric disorder characterized by mood swings (depressive and manic phases) that can strongly affect the quality of life of patients and their families. The lifetime prevalence of BD in the general population is 1%. The pathogenesis of BD is unknown; however, comprehensive epidemiological studies have shown that both genetic and environmental factors play a role. Within the scope of the current project, we aim to determine the genetic change responsible for the emergence of the disease and to make a genotype-phenotype correlation. Methods In this study, we evaluated single nucleotide gene variants in three families (n = 6 patients) with bipolar disorder using whole-exome sequencing. Results Seven genes (TMTC1, DGKH, STARD9, ITIH1, MARCKS, CSMD1, and ADRA2B) were identified as possibly associated with BPD. In addition, two novel variants were presented in the TMTC1 (c.1214T>G) and STARD9 (c.8288C>G) genes. Conclusion Prospective studies in larger patient groups are required to determine the role of these genes in the etiology of the disease and their potential in diagnosis and treatment. To the best of our knowledge, this is the first methodically comprehensive study conducted in our country and can contribute to the identification of genes that may be associated with BD and the etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Esra Çolak-Geniş
- Department of Medical Genetics, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Müjdan Özdemir Erdoğan
- Department of Medical Genetics, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| | - Fethi Sırrı Çam
- Department of Medical Genetics, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Ömer Aydemir
- Department of Psychiatry, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Funda Akin
- Department of Psychiatry, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | | | - Mustafa Solak
- Department of Medical Genetics, Afyonkarahisar University of Health Sciences, Afyonkarahisar, Turkey
| |
Collapse
|
6
|
Capisizu A, Sandu C, Caragea RM, Capisizu AS. A missense mutation in the MACF1 gene in a patient with autism spectrum disorder and epilepsy. J Med Life 2024; 17:1023-1029. [PMID: 39781307 PMCID: PMC11705475 DOI: 10.25122/jml-2024-0312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/28/2024] [Indexed: 01/12/2025] Open
Abstract
The MACF1 gene (OMIM: 608271) encodes the Microtubule-Actin Cross-Linking Factor 1 protein. Existing medical research shows that genetic mutations in the MACF1 gene have been associated with neurodevelopmental and neurodegenerative disorders, with variants of unknown significance also linked to autism spectrum disorder (ASD). However, the number of reported autism disorder or epilepsy cases associated with MACF1 mutations remains limited. We present the case of a 7-year-old girl, a long-term patient at the Pediatric Neurology Clinic of Dr. Alexandru Obregia Hospital in Bucharest, followed since the age of 3. She initially presented with epilepsy characterized by generalized seizures, clinically resembling both spasms and myoclonus. Over time, she exhibited features of a pervasive developmental disorder and moderate cognitive delay. Genetic testing identified a missense point mutation in the MACF1 gene, c.16223C > T, p.(Pro504Leu). Her final diagnosis was epilepsy with generalized seizures of non-lesional origin, moderate cognitive impairment, pervasive developmental disorder, and a confirmed point mutation in the MACF1 gene. This case underscores the importance of incorporating genetic testing into the diagnostic process for patients with autism spectrum disorder and epilepsy.
Collapse
Affiliation(s)
| | - Carmen Sandu
- Department of Pediatric Neurology, Dr. Alexandru Obregia Psychiatry Hospital, Bucharest, Romania
| | | | - Adriana Sorina Capisizu
- Department of Radiology and Imagistic Medicine 1, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
7
|
McGrouther CC, Rangan AV, Di Florio A, Elman JA, Schork NJ, Kelsoe J, Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. Heterogeneity analysis provides evidence for a genetically homogeneous subtype of bipolar-disorder. ARXIV 2024:arXiv:2405.00159v2. [PMID: 38745705 PMCID: PMC11092873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Bipolar Disorder (BD) is a complex disease. It is heterogeneous, both at the phenotypic and genetic level, although the extent and impact of this heterogeneity is not fully understood. One way to assess this heterogeneity is to look for patterns in the subphenotype data. Because of the variability in how phenotypic data was collected by the various BD studies over the years, homogenizing this subphenotypic data is a challenging task, and so is replication. An alternative methodology, taken here, is to set aside the intricacies of subphenotype and allow the genetic data itself to determine which subjects define a homogeneous genetic subgroup (termed 'bicluster' below). Results In this paper, we leverage recent advances in heterogeneity analysis to look for genetically-driven subgroups (i.e., biclusters) within the broad phenotype of Bipolar Disorder. We first apply this covariate-corrected biclustering algorithm to a cohort of 2524 BD cases and 4106 controls from the Bipolar Disease Research Network (BDRN) within the Psychiatric Genomics Consortium (PGC). We find evidence of genetic heterogeneity delineating a statistically significant bicluster comprising a subset of BD cases which exhibits a disease-specific pattern of differential-expression across a subset of SNPs. This disease-specific genetic pattern (i.e., 'genetic subgroup') replicates across the remaining data-sets collected by the PGC containing 5781/8289, 3581/7591, and 6825/9752 cases/controls, respectively. This genetic subgroup (discovered without using any BD subtype information) was more prevalent in Bipolar type-I than in Bipolar type-II. Conclusions Our methodology has successfully identified a replicable homogeneous genetic subgroup of bipolar disorder. This subgroup may represent a collection of correlated genetic risk-factors for BDI. By investigating the subgroup's bicluster-informed polygenic-risk-scoring (PRS), we find that the disease-specific pattern highlighted by the bicluster can be leveraged to eliminate noise from our GWAS analyses and improve risk prediction. This improvement is particularly notable when using only a relatively small subset of the available SNPs, implying improved SNP replication. Though our primary focus is only the analysis of disease-related signal, we also identify replicable control-related heterogeneity.
Collapse
Affiliation(s)
- Caroline C. McGrouther
- Courant Institute of Mathematical Sciences, New York University, New York, NY, United States of America
| | - Aaditya V. Rangan
- Courant Institute of Mathematical Sciences, New York University, New York, NY, United States of America
| | - Arianna Di Florio
- School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Jeremy A. Elman
- Department of Psychiatry, University of California San Diego, San Diego, CA, United States of America
| | - Nicholas J. Schork
- The Translational Genomics Research Institute, Quantitative Medicine and Systems Biology, Phoenix, AZ, United States of America
| | - John Kelsoe
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America
| | | |
Collapse
|
8
|
Agogo-Mawuli PS, Mendez J, Oestreich EA, Bosch DE, Siderovski DP. Molecular Modeling and In Vitro Functional Analysis of the RGS12 PDZ Domain Variant Associated with High-Penetrance Familial Bipolar Disorder. Int J Mol Sci 2024; 25:11431. [PMID: 39518985 PMCID: PMC11546610 DOI: 10.3390/ijms252111431] [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: 08/01/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Bipolar disorder's etiology involves genetics, environmental factors, and gene-environment interactions, underlying its heterogeneous nature and treatment complexity. In 2020, Forstner and colleagues catalogued 378 sequence variants co-segregating with familial bipolar disorder. A notable candidate was an R59Q missense mutation in the PDZ (PSD-95/Dlg1/ZO-1) domain of RGS12. We previously demonstrated that RGS12 loss removes negative regulation on the kappa opioid receptor, disrupting basal ganglia dopamine homeostasis and dampening responses to dopamine-eliciting psychostimulants. Here, we investigated the R59Q variation in the context of potential PDZ domain functional alterations. We first validated a new target for the wildtype RGS12 PDZ domain-the SAPAP3 C-terminus-by molecular docking, surface plasmon resonance (SPR), and co-immunoprecipitation. While initial molecular dynamics (MD) studies predicted negligible effects of the R59Q variation on ligand binding, SPR showed a significant reduction in binding affinity for the three peptide targets tested. AlphaFold2-generated models predicted a modest reduction in protein-peptide interactions, which is consistent with the reduced binding affinity observed by SPR, suggesting that the substituted glutamine side chain may weaken the affinity of RGS12 for its in vivo binding targets, likely through allosteric changes. This difference may adversely affect the CNS signaling related to dynorphin and dopamine in individuals with this R59Q variation, potentially impacting bipolar disorder pathophysiology.
Collapse
Affiliation(s)
- Percy S. Agogo-Mawuli
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| | - Joseph Mendez
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| | - Emily A. Oestreich
- Department of Biomedical Sciences, Pacific Northwest University of Health Sciences, Yakima, WA 98901, USA
| | - Dustin E. Bosch
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - David P. Siderovski
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| |
Collapse
|
9
|
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
|
10
|
Shang B, Yang R, Lian K, Dong L, Liu H, Wang T, Yang G, Xi K, Xu X, Cheng Y. Family-based genetic analysis in schizophrenia by whole-exome sequence to identify rare pathogenic variants. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32968. [PMID: 38293813 DOI: 10.1002/ajmg.b.32968] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 12/29/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
Schizophrenia (SCZ) is influenced by a combination of genetic and environmental factors. Although several studies have been conducted to identify the causative loci and genes, few of these loci or genes can be repeated due to the high phenotypic and genetic heterogeneity of disease, and their mechanisms are not fully understood. There may be some "missing heritability" that has not yet been found. In order to investigate the deleterious heritable mutations, whole-exome sequencing (WES) in pedigrees with SCZ was used in the current work. Two unrelated pedigrees with SCZ were recruited to perform WES. Genetic analysis was next performed to find potential variants in accordance with the prioritized strategy. Followed by genetic analysis to detect candidate variants according to the prioritized strategy. Next, a series of algorithms was used to predict the pathogenicity of variants. Sanger sequencing was finally conducted to verify the co-segregation. Recessive mutations in six genes (TFEB, SNAI2, TFAP2B, PRKDC, ST18 in Pedigree 1 and PKHD1L1 in Pedigree 2) that co-segregated with SCZ in two families were discovered through genetic analysis by WES. Sanger sequencing verified that all of the mutations in the affected siblings were homozygous. These results corroborated the hypothesis that SCZ exhibits strong heterogeneity and complex inheritance patterns. The newly discovered homozygous variations deepen our understanding of the mutation spectrum and offer more proof for the involvement of TFEB, SNAI2, TFAP2B, PRKDC, ST18, and PKHD1L1 in the development of SCZ.
Collapse
Affiliation(s)
- Binli Shang
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Runxu Yang
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan medical Centre for Mental Health, Kunming, China
| | - Kun Lian
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lei Dong
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | | | | | - Kang Xi
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiufeng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan medical Centre for Mental Health, Kunming, China
| | - Yuqi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan medical Centre for Mental Health, Kunming, China
| |
Collapse
|
11
|
Ma T, Zhou S, Xie X, Chen J, Wang J, Zhang G. A case report of a family with developmental arrest of human prokaryotic stage zygote. Front Cell Dev Biol 2024; 12:1280797. [PMID: 38606321 PMCID: PMC11006971 DOI: 10.3389/fcell.2024.1280797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/19/2024] [Indexed: 04/13/2024] Open
Abstract
To study the genetic variation leading to the arrest phenotype of pronuclear (PN) zygotes. We recruited a family characterized by recurrent PN arrest during in vitro fertilization (IVF) and intracytoplasmic sperm injection cycles (ICSI) and performed whole-exome sequencing for 2 individuals. The transcriptome profiles of PN-arrest zygotes were assessed by single-cell RNA sequencing analysis. The variants were then validated by PCR amplification and Sanger sequencing in the affected individuals and other family members. A family characterized by recurrent PN arrest during IVF and ICSI cycles were enrolled after giving written informed consent. Peripheral blood samples were taken for DNA extraction. Three PN-arrest zygotes from patient III-3 were used for single-cell RNA-seq as described. This phenotype was reproduced after multiple cycles of egg retrieval and after trying different fertilization methods and multiple ovulation regimens. The mutant genes of whole exon sequencing were screened and verified. The missense variant c. C1630T (p.R544W) in RGS12 was responsible for a phenotype characterized by paternal transmission. RGS12 controls Ca2+ oscillation, which is required for oocyte activation after fertilization. Single-cell transcriptome profiling of PN-arrest zygotes revealed defective established translation, RNA processing and cell cycle, which explained the failure of complete oocyte activation. Furthermore, we identified proximal genes involved in Ca2+ oscillation-cytostatic factor-anaphase-promoting complex (Ca2+ oscillation-CSF-APC) signaling, including upregulated CaMKII, ORAI1, CDC20, and CDH1 and downregulated EMI1 and BUB3. The findings indicate abnormal spontaneous Ca2+ oscillations leading to oocytes with prolonged low CSF level and high APC level, which resulted in defective nuclear envelope breakdown and DNA replication. We have identified an RGS12 variant as the potential cause of female infertility characterized by arrest at the PN stage during multiple IVF and ICSI.
Collapse
Affiliation(s)
- Tianzhong Ma
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Songxia Zhou
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xuezhen Xie
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jingyao Chen
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jing Wang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
Salem D, Fecek RJ. Role of microtubule actin crosslinking factor 1 (MACF1) in bipolar disorder pathophysiology and potential in lithium therapeutic mechanism. Transl Psychiatry 2023; 13:221. [PMID: 37353479 DOI: 10.1038/s41398-023-02483-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/25/2023] Open
Abstract
Bipolar affective disorder (BPAD) are life-long disorders that account for significant morbidity in afflicted patients. The etiology of BPAD is complex, combining genetic and environmental factors to increase the risk of disease. Genetic studies have pointed toward cytoskeletal dysfunction as a potential molecular mechanism through which BPAD may arise and have implicated proteins that regulate the cytoskeleton as risk factors. Microtubule actin crosslinking factor 1 (MACF1) is a giant cytoskeletal crosslinking protein that can coordinate the different aspects of the mammalian cytoskeleton with a wide variety of actions. In this review, we seek to highlight the functions of MACF1 in the nervous system and the molecular mechanisms leading to BPAD pathogenesis. We also offer a brief perspective on MACF1 and the role it may be playing in lithium's mechanism of action in treating BPAD.
Collapse
Affiliation(s)
- Deepak Salem
- Lake Erie College of Osteopathic Medicine at Seton Hill, Department of Microbiology, Greensburg, USA
- University of Maryland Medical Center/Sheppard Pratt Psychiatry Residency Program, Baltimore, USA
| | - Ronald J Fecek
- Lake Erie College of Osteopathic Medicine at Seton Hill, Department of Microbiology, Greensburg, USA.
| |
Collapse
|
13
|
Medina AM, Hagenauer MH, Krolewski DM, Hughes E, Forrester LCT, Walsh DM, Waselus M, Richardson E, Turner CA, Sequeira PA, Cartagena PM, Thompson RC, Vawter MP, Bunney BG, Myers RM, Barchas JD, Lee FS, Schatzberg AF, Bunney WE, Akil H, Watson SJ. Neurotransmission-related gene expression in the frontal pole is altered in subjects with bipolar disorder and schizophrenia. Transl Psychiatry 2023; 13:118. [PMID: 37031222 PMCID: PMC10082811 DOI: 10.1038/s41398-023-02418-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/10/2023] Open
Abstract
The frontal pole (Brodmann area 10, BA10) is the largest cytoarchitectonic region of the human cortex, performing complex integrative functions. BA10 undergoes intensive adolescent grey matter pruning prior to the age of onset for bipolar disorder (BP) and schizophrenia (SCHIZ), and its dysfunction is likely to underly aspects of their shared symptomology. In this study, we investigated the role of BA10 neurotransmission-related gene expression in BP and SCHIZ. We performed qPCR to measure the expression of 115 neurotransmission-related targets in control, BP, and SCHIZ postmortem samples (n = 72). We chose this method for its high sensitivity to detect low-level expression. We then strengthened our findings by performing a meta-analysis of publicly released BA10 microarray data (n = 101) and identified sources of convergence with our qPCR results. To improve interpretation, we leveraged the unusually large database of clinical metadata accompanying our samples to explore the relationship between BA10 gene expression, therapeutics, substances of abuse, and symptom profiles, and validated these findings with publicly available datasets. Using these convergent sources of evidence, we identified 20 neurotransmission-related genes that were differentially expressed in BP and SCHIZ in BA10. These results included a large diagnosis-related decrease in two important therapeutic targets with low levels of expression, HTR2B and DRD4, as well as other findings related to dopaminergic, GABAergic and astrocytic function. We also observed that therapeutics may produce a differential expression that opposes diagnosis effects. In contrast, substances of abuse showed similar effects on BA10 gene expression as BP and SCHIZ, potentially amplifying diagnosis-related dysregulation.
Collapse
Affiliation(s)
- Adriana M Medina
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - David M Krolewski
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Evan Hughes
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Maria Waselus
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Evelyn Richardson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Cortney A Turner
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Robert C Thompson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | | | | | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Stanley J Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Privitera F, Trusso MA, Valentino F, Doddato G, Fallerini C, Brunelli G, D'Aurizio R, Furini S, Goracci A, Fagiolini A, Mari F, Renieri A, Ariani F. Heterozygosity for neuronal ceroid lipofuscinosis predisposes to bipolar disorder. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45:11-19. [PMID: 35881528 PMCID: PMC9976914 DOI: 10.47626/1516-4446-2022-2650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Bipolar disorder is a heritable chronic mental disorder that causes psychosocial impairment through depressive/manic episodes. Familial transmission of bipolar disorder does not follow simple Mendelian patterns of inheritance. The aim of this study was to describe a large family with 12 members affected by bipolar disorder. Whole-exome sequencing was performed for eight members, three of whom were diagnosed with bipolar disorder, and another reported as "borderline." METHODS Whole-exome sequencing data allowed us to select variants that the affected members had in common, including and excluding the "borderline" individual with moderate anxiety and obsessive-compulsive traits. RESULTS The results favored designating certain genes as predispositional to bipolar disorder: a heterozygous missense variant in CLN6 resulted in a "borderline" phenotype that, if combined with a heterozygous missense variant in ZNF92, is responsible for the more severe bipolar disorder phenotype. Both rare missense changes are predicted to disrupt protein function. CONCLUSIONS Loss of both alleles in CLN6 causes neuronal ceroid lipofuscinosis, a severe progressive childhood neurological disorder. Our results indicate that heterozygous CLN6 carriers, previously reported as healthy, may be susceptible to bipolar disorder later in life if associated with additional variants in ZNF92.
Collapse
Affiliation(s)
- Flavia Privitera
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Maria A Trusso
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Floriana Valentino
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Gabriella Doddato
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Giulia Brunelli
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics, National Research Council, Pisa, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Arianna Goracci
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy. Department of Mental Health; Psychiatry Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Andrea Fagiolini
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Francesca Mari
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Francesca Ariani
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
15
|
Chitre AS, Hebda-Bauer EK, Blandino P, Bimschleger H, Nguyen KM, Maras P, Li F, Ozel AB, Pan Y, Polesskaya O, Cheng R, Flagel SB, Watson SJ, Li J, Akil H, Palmer AA. Genome-wide association study in a rat model of temperament identifies multiple loci for exploratory locomotion and anxiety-like traits. Front Genet 2023; 13:1003074. [PMID: 36712851 PMCID: PMC9873817 DOI: 10.3389/fgene.2022.1003074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 01/12/2023] Open
Abstract
Common genetic factors likely contribute to multiple psychiatric diseases including mood and substance use disorders. Certain stable, heritable traits reflecting temperament, termed externalizing or internalizing, play a large role in modulating vulnerability to these disorders. To model these heritable tendencies, we selectively bred rats for high and low exploration in a novel environment [bred High Responders (bHR) vs. Low Responders (bLR)]. To identify genes underlying the response to selection, we phenotyped and genotyped 538 rats from an F2 cross between bHR and bLR. Several behavioral traits show high heritability, including the selection trait: exploratory locomotion (EL) in a novel environment. There were significant phenotypic and genetic correlations between tests that capture facets of EL and anxiety. There were also correlations with Pavlovian conditioned approach (PavCA) behavior despite the lower heritability of that trait. Ten significant and conditionally independent loci for six behavioral traits were identified. Five of the six traits reflect different facets of EL that were captured by three behavioral tests. Distance traveled measures from the open field and the elevated plus maze map onto different loci, thus may represent different aspects of novelty-induced locomotor activity. The sixth behavioral trait, number of fecal boli, is the only anxiety-related trait mapping to a significant locus on chromosome 18 within which the Pik3c3 gene is located. There were no significant loci for PavCA. We identified a missense variant in the Plekhf1 gene on the chromosome 1:95 Mb QTL and Fancf and Gas2 as potential candidate genes that may drive the chromosome 1:107 Mb QTL for EL traits. The identification of a locomotor activity-related QTL on chromosome 7 encompassing the Pkhd1l1 and Trhr genes is consistent with our previous finding of these genes being differentially expressed in the hippocampus of bHR vs. bLR rats. The strong heritability coupled with identification of several loci associated with exploratory locomotion and emotionality provide compelling support for this selectively bred rat model in discovering relatively large effect causal variants tied to elements of internalizing and externalizing behaviors inherent to psychiatric and substance use disorders.
Collapse
Affiliation(s)
- Apurva S. Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Elaine K. Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Peter Blandino
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Hannah Bimschleger
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Pamela Maras
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Fei Li
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - A. Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Yanchao Pan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Shelly B. Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Stanley J. Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Jun Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States,Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States,*Correspondence: Abraham A. Palmer,
| |
Collapse
|
16
|
Chen K, Ye C, Gao Z, Hu J, Chen C, Xiao R, Lu F, Wei K. Immune infiltration patterns and identification of new diagnostic biomarkers GDF10, NCKAP5, and RTKN2 in non-small cell lung cancer. Transl Oncol 2023; 29:101618. [PMID: 36628881 PMCID: PMC9843486 DOI: 10.1016/j.tranon.2023.101618] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
This study aimed to identify potential biomarkers for non-small cell lung cancer (NSCLC) and analyze the role of immune cell infiltration in NSCLC. R software was used to screen differentially expressed genes (DEGs) from NSCLC datasets obtained from the Gene Expression Omnibus (GEO) database, and functional correlation analysis was performed. The machine learning algorithms were used to screen the potential biomarkers of NSCLC. The diagnostic values were assessed through receiver operating characteristic (ROC) curves. The protein and mRNA expression levels of potential biomarkers were verified based on the Human Protein Atlas (HPA) database and qRT-PCR. CIBERSORT was used to evaluate the infiltration of immune cells in NSCLC tissues, and the correlation between potential biomarkers and infiltrated immune cell was analyzed. Finally, specific siRNAs were utilized to reduce the GDF10, NCKAP5, and RTKN2 expression in A549 and H1975 cells. The proliferation ability of A549 and H1975 cells was detected by MTT assay. A total of 848 upregulated DEGs and 1308 downregulated DEGs were identified. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that the DEGs were mainly related to cell division. Disease ontology (DO) enrichment analysis showed that the diseases with these DEGs were mainly lung diseases, including NSCLC. In addition,three potential biomarkers were identified: GDF10, NCKAP5, and RTKN2. Immune cell infiltration analysis showed that resting NK cells, activated dendritic cells, and Tregs may be involved in the pathogenesis of NSCLC. Meanwhile, GDF10, NCKAP5, and RTKN2 were negatively correlated with Tregs and naïve B cells but were positively correlated with activated dendritic cells and resting NK cells. Immunohistochemical staining showed that the expression of GDF10, NCKAP5, and RTKN2 in the lung tissue of patients with NSCLC was lower than that of normal lung tissue. qRT-PCR also confirmed that the mRNA expression of three biomarkers in NSCLC cell lines A549 and H1975 were significantly lower than those in human normal lung epithelial cells BEAS-2B. An MTT assay showed that GDF10, NCKAP5, and RTKN2 knockdown significantly promoted the proliferation of A549 and H1975 cells. The in vitro experiments showed that GDF10, NCKAP5, and RTKN2 played the inhibitory effects on NSCLC cell lines proliferation. Hence, GDF10, NCKAP5, and RTKN2 can be used as diagnostic biomarkers for NSCLC.
Collapse
|
17
|
Ganesh S, Vemula A, Bhattacharjee S, Mathew K, Ithal D, Navin K, Nadella RK, Viswanath B, Sullivan PF, Jain S, Purushottam M. Whole exome sequencing in dense families suggests genetic pleiotropy amongst Mendelian and complex neuropsychiatric syndromes. Sci Rep 2022; 12:21128. [PMID: 36476812 PMCID: PMC9729597 DOI: 10.1038/s41598-022-25664-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Whole Exome Sequencing (WES) studies provide important insights into the genetic architecture of serious mental illness (SMI). Genes that are central to the shared biology of SMIs may be identified by WES in families with multiple affected individuals with diverse SMI (F-SMI). We performed WES in 220 individuals from 75 F-SMI families and 60 unrelated controls. Within pedigree prioritization employed criteria of rarity, functional consequence, and sharing by ≥ 3 affected members. Across the sample, gene and gene-set-wide case-control association analysis was performed with Sequence Kernel Association Test (SKAT). In 14/16 families with ≥ 3 sequenced affected individuals, we identified a total of 78 rare predicted deleterious variants in 78 unique genes shared by ≥ 3 members with SMI. Twenty (25%) genes were implicated in monogenic CNS syndromes in OMIM (OMIM-CNS), a fraction that is a significant overrepresentation (Fisher's Exact test OR = 2.47, p = 0.001). In gene-set SKAT, statistically significant association was noted for OMIM-CNS gene-set (SKAT-p = 0.005) but not the synaptic gene-set (SKAT-p = 0.17). In this WES study in F-SMI, we identify private, rare, protein altering variants in genes previously implicated in Mendelian neuropsychiatric syndromes; suggesting pleiotropic influences in neurodevelopment between complex and Mendelian syndromes.
Collapse
Affiliation(s)
- Suhas Ganesh
- Central Institute of Psychiatry, Kanke, Ranchi, India
- Schizophrenia Neuropharmacology Research Group, Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | - Alekhya Vemula
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | | | - Kezia Mathew
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | - Dhruva Ithal
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | - Karthick Navin
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | - Ravi Kumar Nadella
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
- Department of Psychiatry, Varma Hospital, Bhimavaram, India
| | - Biju Viswanath
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | - Patrick F Sullivan
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics at Karolinska Institutet, Stockholm, Sweden
| | - Sanjeev Jain
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India
| | - Meera Purushottam
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, India.
| |
Collapse
|
18
|
Pedigree-based study to identify GOLGB1 as a risk gene for bipolar disorder. Transl Psychiatry 2022; 12:390. [PMID: 36115840 PMCID: PMC9482626 DOI: 10.1038/s41398-022-02163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Bipolar disorder (BD) is a complex psychiatric disorder with strong heritability. Identification of new BD risk genes will help determine the mechanism underlying disease pathogenesis. In the present study, we carried out whole genome sequencing for a Chinese BD family with three affected members and three unaffected members, and identified multiple candidate causal variations, including a frameshift mutation in the GOLGB1 gene. Since a GOLGB1 missense mutation was also found in another BD pedigree, we carried out functional studies by downregulating Golgb1 expression in the brain of neonatal mice. Golgb1 deficiency had no effect on anxiety, memory, and social behaviors in young adult mice. However, we found that young adult mice with Golgb1 deficiency exhibited elevated locomotor activity and decreased depressive behaviors in the tail suspension test and the sucrose preference test, but increased depressive behaviors in the forced swim test, resembling the dual character of BD patients with both mania and depression. Moreover, Golgb1 downregulation reduced PSD93 levels and Akt phosphorylation in the brain. Together, our results indicate that GOLGB1 is a strong BD risk gene candidate whose deficiency may result in BD phenotypes possibly through affecting PSD93 and PI3K/Akt signaling.
Collapse
|
19
|
Defining Specific Cell States of MPTP-Induced Parkinson's Disease by Single-Nucleus RNA Sequencing. Int J Mol Sci 2022; 23:ijms231810774. [PMID: 36142685 PMCID: PMC9504791 DOI: 10.3390/ijms231810774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with an impairment of movement execution that is related to age and genetic and environmental factors. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin widely used to induce PD models, but the effect of MPTP on the cells and genes of PD has not been fully elucidated. By single-nucleus RNA sequencing, we uncovered the PD-specific cells and revealed the changes in their cellular states, including astrocytosis and endothelial cells' absence, as well as a cluster of medium spiny neuron cells unique to PD. Furthermore, trajectory analysis of astrocyte and endothelial cell populations predicted candidate target gene sets that might be associated with PD. Notably, the detailed regulatory roles of astrocyte-specific transcription factors Dbx2 and Sox13 in PD were revealed in our work. Finally, we characterized the cell-cell communications of PD-specific cells and found that the overall communication strength was enhanced in PD compared with a matched control, especially the signaling pathways of NRXN and NEGR. Our work provides an overview of the changes in cellular states of the MPTP-induced mouse brain.
Collapse
|
20
|
Roles and mechanisms of ankyrin-G in neuropsychiatric disorders. Exp Mol Med 2022; 54:867-877. [PMID: 35794211 PMCID: PMC9356056 DOI: 10.1038/s12276-022-00798-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022] Open
Abstract
Ankyrin proteins act as molecular scaffolds and play an essential role in regulating cellular functions. Recent evidence has implicated the ANK3 gene, encoding ankyrin-G, in bipolar disorder (BD), schizophrenia (SZ), and autism spectrum disorder (ASD). Within neurons, ankyrin-G plays an important role in localizing proteins to the axon initial segment and nodes of Ranvier or to the dendritic shaft and spines. In this review, we describe the expression patterns of ankyrin-G isoforms, which vary according to the stage of brain development, and consider their functional differences. Furthermore, we discuss how posttranslational modifications of ankyrin-G affect its protein expression, interactions, and subcellular localization. Understanding these mechanisms leads us to elucidate potential pathways of pathogenesis in neurodevelopmental and psychiatric disorders, including BD, SZ, and ASD, which are caused by rare pathogenic mutations or changes in the expression levels of ankyrin-G in the brain. Mutations affecting the production, distribution, or function of the ankyrin-G protein may contribute to a variety of different neuropsychiatric disorders. Ankyrin-G is typically observed at the synapses between neurons, and contributes to intercellular adhesion and signaling along with other important functions. Peter Penzes and colleagues at Northwestern University, Chicago, USA, review the biology of this protein and identify potential mechanisms by which ankyrin-G mutations might impair healthy brain development. Mutations in the gene encoding this protein are strongly linked with bipolar disorder, but have also been tentatively connected to autism spectrum disorders and schizophrenia. The authors highlight physiologically important interactions with a diverse array of other brain proteins, which can in turn be modulated by various chemical modifications to ankyrin-G, and conclude that drugs that influence these modifications could have potential therapeutic value.
Collapse
|
21
|
Li X, Ma S, Yan W, Wu Y, Kong H, Zhang M, Luo X, Xia J. dbBIP: a comprehensive bipolar disorder database for genetic research. Database (Oxford) 2022; 2022:baac049. [PMID: 35779245 PMCID: PMC9250320 DOI: 10.1093/database/baac049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/28/2022] [Accepted: 06/11/2022] [Indexed: 11/17/2022]
Abstract
Bipolar disorder (BIP) is one of the most common hereditary psychiatric disorders worldwide. Elucidating the genetic basis of BIP will play a pivotal role in mechanistic delineation. Genome-wide association studies (GWAS) have successfully reported multiple susceptibility loci conferring BIP risk, thus providing insight into the effects of its underlying pathobiology. However, difficulties remain in the extrication of important and biologically relevant data from genetic discoveries related to psychiatric disorders such as BIP. There is an urgent need for an integrated and comprehensive online database with unified access to genetic and multi-omics data for in-depth data mining. Here, we developed the dbBIP, a database for BIP genetic research based on published data. The dbBIP consists of several modules, i.e.: (i) single nucleotide polymorphism (SNP) module, containing large-scale GWAS genetic summary statistics and functional annotation information relevant to risk variants; (ii) gene module, containing BIP-related candidate risk genes from various sources and (iii) analysis module, providing a simple and user-friendly interface to analyze one's own data. We also conducted extensive analyses, including functional SNP annotation, integration (including summary-data-based Mendelian randomization and transcriptome-wide association studies), co-expression, gene expression, tissue expression, protein-protein interaction and brain expression quantitative trait loci analyses, thus shedding light on the genetic causes of BIP. Finally, we developed a graphical browser with powerful search tools to facilitate data navigation and access. The dbBIP provides a comprehensive resource for BIP genetic research as well as an integrated analysis platform for researchers and can be accessed online at http://dbbip.xialab.info. Database URL: http://dbbip.xialab.info.
Collapse
Affiliation(s)
- Xiaoyan Li
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Shunshuai Ma
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Wenhui Yan
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Yong Wu
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, 93 Youyi Road, Qiaokou District, Wuhan, Hubei 430030, China
| | - Hui Kong
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Mingshan Zhang
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| | - Xiongjian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiaochang East Road, Wuhua District, Kunming, Yunnan 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, 19 Qingsong Road, Panlong District, Kunming, Yunnan 650204, China
| | - Junfeng Xia
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education and Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Shushan District, Hefei, Anhui 230601, China
| |
Collapse
|
22
|
Abstract
BACKGROUND To date, besides genome-wide association studies, a variety of other genetic analyses (e.g. polygenic risk scores, whole-exome sequencing and whole-genome sequencing) have been conducted, and a large amount of data has been gathered for investigating the involvement of common, rare and very rare types of DNA sequence variants in bipolar disorder. Also, non-invasive neuroimaging methods can be used to quantify changes in brain structure and function in patients with bipolar disorder. AIMS To provide a comprehensive assessment of genetic findings associated with bipolar disorder, based on the evaluation of different genomic approaches and neuroimaging studies. METHOD We conducted a PubMed search of all relevant literatures from the beginning to the present, by querying related search strings. RESULTS ANK3, CACNA1C, SYNE1, ODZ4 and TRANK1 are five genes that have been replicated as key gene candidates in bipolar disorder pathophysiology, through the investigated studies. The percentage of phenotypic variance explained by the identified variants is small (approximately 4.7%). Bipolar disorder polygenic risk scores are associated with other psychiatric phenotypes. The ENIGMA-BD studies show a replicable pattern of lower cortical thickness, altered white matter integrity and smaller subcortical volumes in bipolar disorder. CONCLUSIONS The low amount of explained phenotypic variance highlights the need for further large-scale investigations, especially among non-European populations, to achieve a more complete understanding of the genetic architecture of bipolar disorder and the missing heritability. Combining neuroimaging data with genetic data in large-scale studies might help researchers acquire a better knowledge of the engaged brain regions in bipolar disorder.
Collapse
Affiliation(s)
- Mojtaba Oraki Kohshour
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Iran
| | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Psychiatry and Psychotherapy, University Hospital LMU Munich, Germany
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, USA
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics, University Hospital LMU Munich, Germany; and Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, USA
| |
Collapse
|
23
|
O'Connell KS, Coombes BJ. Genetic contributions to bipolar disorder: current status and future directions. Psychol Med 2021; 51:2156-2167. [PMID: 33879273 PMCID: PMC8477227 DOI: 10.1017/s0033291721001252] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/12/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a highly heritable mental disorder and is estimated to affect about 50 million people worldwide. Our understanding of the genetic etiology of BD has greatly increased in recent years with advances in technology and methodology as well as the adoption of international consortiums and large population-based biobanks. It is clear that BD is also highly heterogeneous and polygenic and shows substantial genetic overlap with other psychiatric disorders. Genetic studies of BD suggest that the number of associated loci is expected to substantially increase in larger future studies and with it, improved genetic prediction of the disorder. Still, a number of challenges remain to fully characterize the genetic architecture of BD. First among these is the need to incorporate ancestrally-diverse samples to move research away from a Eurocentric bias that has the potential to exacerbate health disparities already seen in BD. Furthermore, incorporation of population biobanks, registry data, and electronic health records will be required to increase the sample size necessary for continued genetic discovery, while increased deep phenotyping is necessary to elucidate subtypes within BD. Lastly, the role of rare variation in BD remains to be determined. Meeting these challenges will enable improved identification of causal variants for the disorder and also allow for equitable future clinical applications of both genetic risk prediction and therapeutic interventions.
Collapse
Affiliation(s)
- Kevin S. O'Connell
- Division of Mental Health and Addiction, NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, 0407Oslo, Norway
| | - Brandon J. Coombes
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
24
|
Pol-Fuster J, Cañellas F, Ruiz-Guerra L, Medina-Dols A, Bisbal-Carrió B, Ortega-Vila B, Llinàs J, Hernandez-Rodriguez J, Lladó J, Olmos G, Strauch K, Heine-Suñer D, Vives-Bauzà C, Flaquer A. The conserved ASTN2/BRINP1 locus at 9q33.1-33.2 is associated with major psychiatric disorders in a large pedigree from Southern Spain. Sci Rep 2021; 11:14529. [PMID: 34267256 PMCID: PMC8282839 DOI: 10.1038/s41598-021-93555-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/21/2021] [Indexed: 11/11/2022] Open
Abstract
We investigated the genetic causes of major mental disorders (MMDs) including schizophrenia, bipolar disorder I, major depressive disorder and attention deficit hyperactive disorder, in a large family pedigree from Alpujarras, South of Spain, a region with high prevalence of psychotic disorders. We applied a systematic genomic approach based on karyotyping (n = 4), genotyping by genome-wide SNP array (n = 34) and whole-genome sequencing (n = 12). We performed genome-wide linkage analysis, family-based association analysis and polygenic risk score estimates. Significant linkage was obtained at chromosome 9 (9q33.1–33.2, LOD score = 4.11), a suggestive region that contains five candidate genes ASTN2, BRINP1, C5, TLR4 and TRIM32, previously associated with MMDs. Comprehensive analysis associated the MMD phenotype with genes of the immune system with dual brain functions. Moreover, the psychotic phenotype was enriched for genes involved in synapsis. These results should be considered once studying the genetics of psychiatric disorders in other families, especially the ones from the same region, since founder effects may be related to the high prevalence.
Collapse
Affiliation(s)
- Josep Pol-Fuster
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain.,Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Francesca Cañellas
- Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain.,Department of Psychiatry, HUSE, IdISBa, Palma, Spain
| | - Laura Ruiz-Guerra
- Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Aina Medina-Dols
- Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Bàrbara Bisbal-Carrió
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain.,Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Bernat Ortega-Vila
- Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain.,Molecular Diagnostics and Clinical Genetics Unit (UDMGC) and Genomics of Health Research Group, Hospital Universitari Son Espases (HUSE) and Institut d'Investigacions Sanitaries de Balears (IDISBA), Palma, Spain
| | - Jaume Llinàs
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Jessica Hernandez-Rodriguez
- Molecular Diagnostics and Clinical Genetics Unit (UDMGC) and Genomics of Health Research Group, Hospital Universitari Son Espases (HUSE) and Institut d'Investigacions Sanitaries de Balears (IDISBA), Palma, Spain
| | - Jerònia Lladó
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain.,Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Gabriel Olmos
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain.,Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany.,Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, LMU Munich, Munich, Germany
| | - Damià Heine-Suñer
- Molecular Diagnostics and Clinical Genetics Unit (UDMGC) and Genomics of Health Research Group, Hospital Universitari Son Espases (HUSE) and Institut d'Investigacions Sanitaries de Balears (IDISBA), Palma, Spain
| | - Cristòfol Vives-Bauzà
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain. .,Neurobiology Laboratory, Research Unit, Son Espases University Hospital (HUSE), Health Research Institute of Balearic Islands (IdISBa), Floor -1, Module F, R-805, Palma, Spain.
| | - Antònia Flaquer
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany.,Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, LMU Munich, Munich, Germany
| |
Collapse
|
25
|
Clinical and genetic differences between bipolar disorder type 1 and 2 in multiplex families. Transl Psychiatry 2021; 11:31. [PMID: 33431802 PMCID: PMC7801527 DOI: 10.1038/s41398-020-01146-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 01/29/2023] Open
Abstract
The two major subtypes of bipolar disorder (BD), BD-I and BD-II, are distinguished based on the presence of manic or hypomanic episodes. Historically, BD-II was perceived as a less severe form of BD-I. Recent research has challenged this concept of a severity continuum. Studies in large samples of unrelated patients have described clinical and genetic differences between the subtypes. Besides an increased schizophrenia polygenic risk load in BD-I, these studies also observed an increased depression risk load in BD-II patients. The present study assessed whether such clinical and genetic differences are also found in BD patients from multiplex families, which exhibit reduced genetic and environmental heterogeneity. Comparing 252 BD-I and 75 BD-II patients from the Andalusian Bipolar Family (ABiF) study, the clinical course, symptoms during depressive and manic episodes, and psychiatric comorbidities were analyzed. Furthermore, polygenic risk scores (PRS) for BD, schizophrenia, and depression were assessed. BD-I patients not only suffered from more severe symptoms during manic episodes but also more frequently showed incapacity during depressive episodes. A higher BD PRS was significantly associated with suicidal ideation. Moreover, BD-I cases exhibited lower depression PRS. In line with a severity continuum from BD-II to BD-I, our results link BD-I to a more pronounced clinical presentation in both mania and depression and indicate that the polygenic risk load of BD predisposes to more severe disorder characteristics. Nevertheless, our results suggest that the genetic risk burden for depression also shapes disorder presentation and increases the likelihood of BD-II subtype development.
Collapse
|
26
|
Zhang C, Xiao X, Li T, Li M. Translational genomics and beyond in bipolar disorder. Mol Psychiatry 2021; 26:186-202. [PMID: 32424235 DOI: 10.1038/s41380-020-0782-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
Genome-wide association studies (GWAS) have revealed multiple genomic loci conferring risk of bipolar disorder (BD), providing hints for its underlying pathobiology. However, there are still remaining questions to answer. For example, discordance exists between BD heritability estimated with earlier epidemiological evidence and that calculated based on common GWAS variations. Where is the "missing heritability"? How can we explain the biology of the disease based on genetic findings? In this review, we summarize the accomplishments and limitations of current BD GWAS, and discuss potential reasons for the "missing heritability." In addition, progresses of research for the biological mechanisms underlying BD genetic risk using brain tissues, reprogrammed cells, and model animals are reviewed. While our knowledge of BD genetic basis is significantly promoted by these efforts, the complexities of gene regulation in the genome, the spatial-temporal heterogeneity during brain development, and the limitations of different experimental models should always be considered. Notably, several genes have been widely studied given their relatively well-characterized involvement in BD (e.g., CACAN1C and ANK3), and findings of these genes are summarized to both outline possible biological mechanisms of BD and describe examples of translating GWAS discoveries into the pathophysiology.
Collapse
Affiliation(s)
- Chen Zhang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tao Li
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China. .,West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
27
|
Engelbrecht HR, Dalvie S, Agenbag G, Stein DJ, Ramesar RS. Whole-exome sequencing in an Afrikaner family with bipolar disorder. J Affect Disord 2020; 276:69-75. [PMID: 32697718 DOI: 10.1016/j.jad.2020.06.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 06/04/2020] [Accepted: 06/16/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Bipolar disorder (BD) has considerable heritability, with genome-wide association studies indicating that multiple common genetic variants contribute to risk. Less work has been undertaken to assess the contribution of rare variation in the development of this complex disorder, particularly in isolated populations. Using whole-exome sequencing (WES), the aim of this study was to identify rare, potentially damaging variants contributing to risk for BD in the Afrikaner population. METHODS WES was performed on eight Afrikaner family members, five affected and three unaffected. The analyses focused on i) the identification of rare, damaging variation, and ii) the molecular pathways in which these rare variants play a role using in silico prediction tools such as wANNOVAR and KOBAS 3.0. RESULTS Two rare and potentially damaging missense variants in FAM71B and SLC26A9 were shared by affected family members but were absent in unaffected members. In addition, variants in genes that play a role in pathways involved in signal transduction and synaptic transmission were shared by the five affected individuals. LIMITATIONS Two main limitations affect this study: the limited number of cases and controls, and the fact that whole-exome sequencing can only capture a small fragment of the genome which may harbor mutations. CONCLUSION This is the first WES study of BD in an Afrikaner family, and findings suggest that novel candidate genes may contribute to risk for BD in this population. Future work in larger samples of this population as well as in other populations is needed to fully investigate the role of the candidate genes found here.
Collapse
Affiliation(s)
- Hannah-Ruth Engelbrecht
- SA MRC Research Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, 7925.
| | - Shareefa Dalvie
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town.
| | - Gloudi Agenbag
- SA MRC Research Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, 7925.
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town.
| | - Raj S Ramesar
- SA MRC Research Unit for Genomic and Precision Medicine, Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, 7925.
| |
Collapse
|