1
|
Rayan NA, Aow J, Lim MGL, Arcego DM, Ryan R, Nourbakhsh N, de Lima RMS, Craig K, Zhang TY, Goh YT, Sun AX, Tompkins T, Bronner S, Binda S, Diorio J, Parent C, Meaney MJ, Prabhakar S. Shared and unique transcriptomic signatures of antidepressant and probiotics action in the mammalian brain. Mol Psychiatry 2024; 29:3653-3668. [PMID: 38844534 DOI: 10.1038/s41380-024-02619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 11/08/2024]
Abstract
Understanding the shared and divergent mechanisms across antidepressant (AD) classes and probiotics is critical for improving treatment for mood disorders. Here we examine the transcriptomic effects of bupropion (NDRI), desipramine (SNRI), fluoxetine (SSRI) and a probiotic formulation (Lacidofil®) on 10 regions across the mammalian brain. These treatments massively alter gene expression (on average, 2211 differentially expressed genes (DEGs) per region-treatment combination), highlighting the biological complexity of AD and probiotic action. Intersection of DEG sets against neuropsychiatric GWAS loci, sex-specific transcriptomic portraits of major depressive disorder (MDD), and mouse models of stress and depression reveals significant similarities and differences across treatments. Interestingly, molecular responses in the infralimbic cortex, basolateral amygdala and locus coeruleus are region-specific and highly similar across treatments, whilst responses in the Raphe, medial preoptic area, cingulate cortex, prelimbic cortex and ventral dentate gyrus are predominantly treatment-specific. Mechanistically, ADs concordantly downregulate immune pathways in the amygdala and ventral dentate gyrus. In contrast, protein synthesis, metabolism and synaptic signaling pathways are axes of variability among treatments. We use spatial transcriptomics to further delineate layer-specific molecular pathways and DEGs within the prefrontal cortex. Our study reveals complex AD and probiotics action on the mammalian brain and identifies treatment-specific cellular processes and gene targets associated with mood disorders.
Collapse
Affiliation(s)
- Nirmala Arul Rayan
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Jonathan Aow
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Gek Liang Lim
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Richard Ryan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Nooshin Nourbakhsh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | | | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Tie Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Alfred Xuyang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857, Singapore
| | - Thomas Tompkins
- Lallemand Bio-Ingredients, 1620 Rue Prefontaine, Montréal, QC, H1W 2N8, Canada
| | - Stéphane Bronner
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Sylvie Binda
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Carine Parent
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada.
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.
- Brain-Body Initiative, Institute for Cell & Molecular Biology, A*STAR, Singapore, Singapore.
| | - Shyam Prabhakar
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
2
|
Szewczyk LM, Lipiec MA, Liszewska E, Meyza K, Urban-Ciecko J, Kondrakiewicz L, Goncerzewicz A, Rafalko K, Krawczyk TG, Bogaj K, Vainchtein ID, Nakao-Inoue H, Puscian A, Knapska E, Sanders SJ, Jan Nowakowski T, Molofsky AV, Wisniewska MB. Astrocytic β-catenin signaling via TCF7L2 regulates synapse development and social behavior. Mol Psychiatry 2024; 29:57-73. [PMID: 37798419 PMCID: PMC11078762 DOI: 10.1038/s41380-023-02281-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/07/2023]
Abstract
The Wnt/β-catenin pathway contains multiple high-confidence risk genes that are linked to neurodevelopmental disorders, including autism spectrum disorder. However, its ubiquitous roles across brain cell types and developmental stages have made it challenging to define its impact on neural circuit development and behavior. Here, we show that TCF7L2, which is a key transcriptional effector of the Wnt/β-catenin pathway, plays a cell-autonomous role in postnatal astrocyte maturation and impacts adult social behavior. TCF7L2 was the dominant Wnt effector that was expressed in both mouse and human astrocytes, with a peak during astrocyte maturation. The conditional knockout of Tcf7l2 in postnatal astrocytes led to an enlargement of astrocytes with defective tiling and gap junction coupling. These mice also exhibited an increase in the number of cortical excitatory and inhibitory synapses and a marked increase in social interaction by adulthood. These data reveal an astrocytic role for developmental Wnt/β-catenin signaling in restricting excitatory synapse numbers and regulating adult social behavior.
Collapse
Affiliation(s)
- Lukasz Mateusz Szewczyk
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland.
| | - Marcin Andrzej Lipiec
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Liszewska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ksenia Meyza
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Urban-Ciecko
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ludwika Kondrakiewicz
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Goncerzewicz
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Karolina Bogaj
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ilia Davidovich Vainchtein
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Johnson & Johnson, Neuroscience Therapeutic Area, San Diego, CA, USA
| | - Hiromi Nakao-Inoue
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Alicja Puscian
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewelina Knapska
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, OX3 7TY, UK
- New York Genome Center, New York, NY, USA
| | - Tomasz Jan Nowakowski
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Anna Victoria Molofsky
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| | - Marta Barbara Wisniewska
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
3
|
Qi H, Luo L, Lu C, Chen R, Zhou X, Zhang X, Jia Y. TCF7L2 acts as a molecular switch in midbrain to control mammal vocalization through its DNA binding domain but not transcription activation domain. Mol Psychiatry 2023; 28:1703-1717. [PMID: 36782064 DOI: 10.1038/s41380-023-01993-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 01/15/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023]
Abstract
Vocalization is an essential medium for social signaling in birds and mammals. Periaqueductal gray (PAG) a conserved midbrain structure is believed to be responsible for innate vocalizations, but its molecular regulation remains largely unknown. Here, through a mouse forward genetic screening we identified one of the key Wnt/β-catenin effectors TCF7L2/TCF4 controls ultrasonic vocalization (USV) production and syllable complexity during maternal deprivation and sexual encounter. Early developmental expression of TCF7L2 in PAG excitatory neurons is necessary for the complex trait, while TCF7L2 loss reduces neuronal gene expressions and synaptic transmission in PAG. TCF7L2-mediated vocal control is independent of its β-catenin-binding domain but dependent of its DNA binding ability. Patient mutations associated with developmental disorders, including autism spectrum disorders, disrupt the transcriptional repression effect of TCF7L2, while mice carrying those mutations display severe USV impairments. Therefore, we conclude that TCF7L2 orchestrates gene expression in midbrain to control vocal production through its DNA binding but not transcription activation domain.
Collapse
Affiliation(s)
- Huihui Qi
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Medicine, Tsinghua University, Beijing, 100084, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Li Luo
- Tsinghua Laboratory of Brain and Intelligence (THBI), Tsinghua University, Beijing, 100084, China
| | - Caijing Lu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Runze Chen
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Xianyao Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Science, Beijing Normal University, Beijing, 100875, China
| | - Yichang Jia
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China. .,School of Medicine, Tsinghua University, Beijing, 100084, China. .,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China. .,Tsinghua Laboratory of Brain and Intelligence (THBI), Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
4
|
Yang Y, Shen M, Li L, Long Y, Wang L, Lang B, Wu R. Olanzapine Promotes the Occurrence of Metabolic Disorders in Conditional TCF7L2-Knockout Mice. Front Cell Dev Biol 2022; 10:890472. [PMID: 35874808 PMCID: PMC9298277 DOI: 10.3389/fcell.2022.890472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
Objectives: Schizophrenia (SCZ) patients display higher incidence of metabolic syndrome (MetS) and comorbidity of type II diabetes. Both atypical antipsychotics and genetic variants are believed to predispose the patients with the risk, but their interplay remains largely unknown. TCF7L2 is one of the most common genes strongly associated with glucose homeostasis which also participates in the pathogenesis of schizophrenia. In this study, we aimed to explore the regulatory roles of TCF7L2 in atypical antipsychotics-induced MetS. Methods: Mice with pancreatic β-cell–specific Tcf7l2 deletion (CKO) were generated. The CKO mice and control littermates were subjected to olanzapine (4 mg/kg/day) or saline gavage for 6 weeks. Metabolic indices, β cell mass, and the expressing levels of TCF7L2 and GLP-1R in the pancreatic tissue were closely monitored. Results: Tcf7l2 CKO mice displayed a spectrum of core features of MetS, which included remarkably increased rate of weight gain, higher fasting insulin, higher values of blood lipids (cholesterol, triglyceride, and low-density lipoprotein), impaired glucose tolerance, and hypertrophy of adipocytes. Notably, these effects could be further exacerbated by olanzapine. In addition, Tcf7l2 CKO mice with the olanzapine group showed significantly decreased expressions of GLP-1R protein and a trend of reduced pancreatic β-cell mass. RT-qPCR revealed that the CKO mice presented a significantly less transcription of Sp5, an important element of the Wnt signaling pathway. Conclusion: Our study illustrates that mice with pancreatic β-cell–targeted Tcf7l2 deletion were more vulnerable to suffer metabolic abnormalities after olanzapine administration. This impairment may be mediated by the reduced expression of GLP-1R.
Collapse
Affiliation(s)
- Ye Yang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Manjun Shen
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Psychiatry, Shenzhen, China
| | - Li Li
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yujun Long
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lu Wang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bing Lang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bing Lang, ; Renrong Wu,
| | - Renrong Wu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bing Lang, ; Renrong Wu,
| |
Collapse
|
5
|
Seward CH, Saul MC, Troy JM, Dibaeinia P, Zhang H, Sinha S, Stubbs LJ. An epigenomic shift in amygdala marks the transition to maternal behaviors in alloparenting virgin female mice. PLoS One 2022; 17:e0263632. [PMID: 35192674 PMCID: PMC8863255 DOI: 10.1371/journal.pone.0263632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/23/2022] [Indexed: 11/25/2022] Open
Abstract
Adults of many species will care for young offspring that are not their own, a phenomenon called alloparenting. However, in many cases, nonparental adults must be sensitized by repeated or extended exposures to newborns before they will robustly display parental-like behaviors. To capture neurogenomic events underlying the transition to active parental caring behaviors, we analyzed brain gene expression and chromatin profiles of virgin female mice co-housed with pregnant dams during pregnancy and after birth. After an initial display of antagonistic behaviors and a surge of defense-related gene expression, we observed a dramatic shift in the chromatin landscape specifically in amygdala of the pup-exposed virgin females compared to females co-housed with mother before birth, accompanied by a dampening of anxiety-related gene expression. This epigenetic shift coincided with hypothalamic expression of the oxytocin gene and the emergence of behaviors and gene expression patterns classically associated with maternal care. The results outline a neurogenomic program associated with dramatic behavioral changes and suggest molecular networks relevant to human postpartum mental health.
Collapse
Affiliation(s)
- Christopher H. Seward
- Pacific Northwest Research Institute, Seattle, WA, United States of America
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
| | - Michael C. Saul
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
| | - Joseph M. Troy
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
| | - Payam Dibaeinia
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Huimin Zhang
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Lisa J. Stubbs
- Pacific Northwest Research Institute, Seattle, WA, United States of America
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States of America
| |
Collapse
|
6
|
Traniello IM, Robinson GE. Neural and Molecular Mechanisms of Biological Embedding of Social Interactions. Annu Rev Neurosci 2021; 44:109-128. [PMID: 34236891 DOI: 10.1146/annurev-neuro-092820-012959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animals operate in complex environments, and salient social information is encoded in the nervous system and then processed to initiate adaptive behavior. This encoding involves biological embedding, the process by which social experience affects the brain to influence future behavior. Biological embedding is an important conceptual framework for understanding social decision-making in the brain, as it encompasses multiple levels of organization that regulate how information is encoded and used to modify behavior. The framework we emphasize here is that social stimuli provoke short-term changes in neural activity that lead to changes in gene expression on longer timescales. This process, simplified-neurons are for today and genes are for tomorrow-enables the assessment of the valence of a social interaction, an appropriate and rapid response, and subsequent modification of neural circuitry to change future behavioral inclinations in anticipation of environmental changes. We review recent research on the neural and molecular basis of biological embedding in the context of social interactions, with a special focus on the honeybee.
Collapse
Affiliation(s)
- Ian M Traniello
- Neuroscience Program and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA;
| | - Gene E Robinson
- Neuroscience Program and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA; .,Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
7
|
Ganmore I, Livny A, Ravona-Springer R, Cooper I, Alkelai A, Shelly S, Tsarfaty G, Heymann A, Schnaider Beeri M, Greenbaum L. TCF7L2 polymorphisms are associated with amygdalar volume in elderly individuals with Type 2 Diabetes. Sci Rep 2019; 9:15818. [PMID: 31676834 PMCID: PMC6825182 DOI: 10.1038/s41598-019-48899-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/08/2019] [Indexed: 02/06/2023] Open
Abstract
The association between several Single Nucleotide Polymorphisms (SNPs) within the transcription factor 7-like 2 (TCF7L2) gene and Type 2 Diabetes (T2D) as well as additional T2D-related traits is well established. Since alteration in total and regional brain volumes are consistent findings among T2D individuals, we studied the association of four T2D susceptibility SNPS within TCF7L2 (rs7901695, rs7903146, rs11196205, and rs12255372) with volumes of white matter hyperintensities (WMH), gray matter, and regional volumes of amygdala and hippocampus obtained from structural MRI among 191 T2D elderly Jewish individuals. Under recessive genetic model (controlling for age, sex and intracranial volume), we found that for all four SNPs, carriers of two copies of the T2D risk allele (homozygous genotype) had significantly smaller amygdalar volume: rs7901695- CC genotype vs. CT + TT genotypes, p = 0.002; rs7903146-TT vs. TC + CC, p = 0.003; rs11196205- CC vs. CG + GG, p = 0.0003; and rs12255372- TT vs. TG + GG, p = 0.003. Adjusting also for T2D-related covariates, body mass index (BMI), and ancestry did not change the results substantively (rs7901695, p = 0.003; rs7903146, p = 0.005; rs11196205, p = 0.001; and rs12255372, p = 0.005). Conditional analysis demonstrated that only rs11196205 was independently associated with amygdalar volume at a significant level. Separate analysis of left and right amygdala revealed stronger results for left amygdalar volume. Taken together, we report association of TCF7L2 SNPs with amygdalar volume among T2D elderly Jewish patients. Further studies in other populations are required to support these findings and reach more definitive conclusions.
Collapse
Affiliation(s)
- Ithamar Ganmore
- Department of Neurology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel. .,The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel. .,Memory clinic, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Abigail Livny
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ramit Ravona-Springer
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Memory clinic, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Anna Alkelai
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Shahar Shelly
- Department of Neurology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Galia Tsarfaty
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anthony Heymann
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Maccabi Healthcare Services, Tel Aviv, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lior Greenbaum
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| |
Collapse
|
8
|
Bem J, Brożko N, Chakraborty C, Lipiec MA, Koziński K, Nagalski A, Szewczyk ŁM, Wiśniewska MB. Wnt/β-catenin signaling in brain development and mental disorders: keeping TCF7L2 in mind. FEBS Lett 2019; 593:1654-1674. [PMID: 31218672 PMCID: PMC6772062 DOI: 10.1002/1873-3468.13502] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/12/2022]
Abstract
Canonical Wnt signaling, which is transduced by β-catenin and lymphoid enhancer factor 1/T cell-specific transcription factors (LEF1/TCFs), regulates many aspects of metazoan development and tissue renewal. Although much evidence has associated canonical Wnt/β-catenin signaling with mood disorders, the mechanistic links are still unknown. Many components of the canonical Wnt pathway are involved in cellular processes that are unrelated to classical canonical Wnt signaling, thus further blurring the picture. The present review critically evaluates the involvement of classical Wnt/β-catenin signaling in developmental processes that putatively underlie the pathology of mental illnesses. Particular attention is given to the roles of LEF1/TCFs, which have been discussed surprisingly rarely in this context. Highlighting recent discoveries, we propose that alterations in the activity of LEF1/TCFs, and particularly of transcription factor 7-like 2 (TCF7L2), result in defects previously associated with neuropsychiatric disorders, including imbalances in neurogenesis and oligodendrogenesis, the functional disruption of thalamocortical circuitry and dysfunction of the habenula.
Collapse
Affiliation(s)
- Joanna Bem
- Centre of New TechnologiesUniversity of WarsawPoland
| | - Nikola Brożko
- Centre of New TechnologiesUniversity of WarsawPoland
| | | | | | | | | | | | | |
Collapse
|
9
|
Bem J, Brożko N, Chakraborty C, Lipiec MA, Koziński K, Nagalski A, Szewczyk ŁM, Wiśniewska MB. Wnt/β-catenin signaling in brain development and mental disorders: keeping TCF7L2 in mind. FEBS Lett 2019. [PMID: 31218672 DOI: 10.1002/1873−3468.13502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Canonical Wnt signaling, which is transduced by β-catenin and lymphoid enhancer factor 1/T cell-specific transcription factors (LEF1/TCFs), regulates many aspects of metazoan development and tissue renewal. Although much evidence has associated canonical Wnt/β-catenin signaling with mood disorders, the mechanistic links are still unknown. Many components of the canonical Wnt pathway are involved in cellular processes that are unrelated to classical canonical Wnt signaling, thus further blurring the picture. The present review critically evaluates the involvement of classical Wnt/β-catenin signaling in developmental processes that putatively underlie the pathology of mental illnesses. Particular attention is given to the roles of LEF1/TCFs, which have been discussed surprisingly rarely in this context. Highlighting recent discoveries, we propose that alterations in the activity of LEF1/TCFs, and particularly of transcription factor 7-like 2 (TCF7L2), result in defects previously associated with neuropsychiatric disorders, including imbalances in neurogenesis and oligodendrogenesis, the functional disruption of thalamocortical circuitry and dysfunction of the habenula.
Collapse
Affiliation(s)
- Joanna Bem
- Centre of New Technologies, University of Warsaw, Poland
| | - Nikola Brożko
- Centre of New Technologies, University of Warsaw, Poland
| | | | | | - Kamil Koziński
- Centre of New Technologies, University of Warsaw, Poland
| | | | | | | |
Collapse
|
10
|
Chater-Diehl E, Sokolowski D, Alberry B, Singh SM. Coordinated Tcf7l2 regulation in a mouse model implicates Wnt signaling in fetal alcohol spectrum disorders. Biochem Cell Biol 2018; 97:375-379. [PMID: 30398926 DOI: 10.1139/bcb-2018-0215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mouse models of fetal alcohol spectrum disorders (FASD) have repeatedly identified genes with long-term changes in expression, DNA methylation, noncoding RNA, and histone modifications in response to neurodevelopmental alcohol exposure. Articulation of FASD is achieved via alcohol's effect on gene expression, likely involving epigenetic regulation. The list of genes affected is large and heterogeneous, depending on experimental protocol. We present reanalysis and synthesis of results highlighting the Wnt transcription factor 7 like 2 (Tcf7l2) gene as uniquely compatible with hippocampal DNA methylation, histone modifications, and gene expression changes in a coordinated response to neurodevelopmental alcohol exposure. We data-mined the literature for Tcf7l2 alterations in response to prenatal alcohol exposure. Four studies identified changes in brain Tcf7l2 expression in different FASD models. Further, we performed an in silico TCF7L2 binding site analysis for FASD mouse model data sets. Seven of these published gene lists were significantly enriched for TCF7L2 binding, indicating potential functional relationships. Finally, TCF7L2 is involved in regulation of hundreds of genes, with a role in brain development, myelination, and neuronal function. Tcf7l2 may be involved in neurological defects associated with alcohol exposure via dysregulation of many genes through Wnt signaling. Further functional work is warranted to validate this model for FASD.
Collapse
Affiliation(s)
- Eric Chater-Diehl
- Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada
| | - Dustin Sokolowski
- Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada
| | - Bonnie Alberry
- Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada.,Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada
| | - Shiva M Singh
- Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada.,Molecular Genetics Unit, Department of Biology, The University of Western Ontario, London, ON N6A 3K7, Canada
| |
Collapse
|
11
|
Misztal K, Brozko N, Nagalski A, Szewczyk LM, Krolak M, Brzozowska K, Kuznicki J, Wisniewska MB. TCF7L2 mediates the cellular and behavioral response to chronic lithium treatment in animal models. Neuropharmacology 2016; 113:490-501. [PMID: 27793772 DOI: 10.1016/j.neuropharm.2016.10.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 11/15/2022]
Abstract
The mechanism of lithium's therapeutic action remains obscure, hindering the discovery of safer treatments for bipolar disorder. Lithium can act as an inhibitor of the kinase GSK3α/β, which in turn negatively regulates β-catenin, a co-activator of LEF1/TCF transcription factors. However, unclear is whether therapeutic levels of lithium activate β-catenin in the brain, and whether this activation could have a therapeutic significance. To address this issue we chronically treated mice with lithium. Although the level of non-phospho-β-catenin increased in all of the brain areas examined, β-catenin translocated into cellular nuclei only in the thalamus. Similar results were obtained when thalamic and cortical neurons were treated with a therapeutically relevant concentration of lithium in vitro. We tested if TCF7L2, a member of LEF1/TCF family that is highly expressed in the thalamus, facilitated the activation of β-catenin. Silencing of Tcf7l2 in thalamic neurons prevented β-catenin from entering the nucleus, even when the cells were treated with lithium. Conversely, when Tcf7l2 was ectopically expressed in cortical neurons, β-catenin shifted to the nucleus, and lithium augmented this process. Lastly, we silenced tcf7l2 in zebrafish and exposed them to lithium for 3 days, to evaluate whether TCF7L2 is involved in the behavioral response. Lithium decreased the dark-induced activity of control zebrafish, whereas the activity of zebrafish with tcf7l2 knockdown was unaltered. We conclude that therapeutic levels of lithium activate β-catenin selectively in thalamic neurons. This effect is determined by the presence of TCF7L2, and potentially contributes to the therapeutic response.
Collapse
Affiliation(s)
- Katarzyna Misztal
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland
| | - Nikola Brozko
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland; University of Warsaw, Centre of New Technologies, Laboratory of Molecular Neurobiology, Poland; Postgraduate School of Molecular Medicine, Warsaw, Poland
| | - Andrzej Nagalski
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland; University of Warsaw, Centre of New Technologies, Laboratory of Molecular Neurobiology, Poland
| | - Lukasz M Szewczyk
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland; Postgraduate School of Molecular Medicine, Warsaw, Poland
| | - Marta Krolak
- University of Warsaw, College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, Poland
| | - Katarzyna Brzozowska
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland; University of Warsaw, Centre of New Technologies, Laboratory of Molecular Neurobiology, Poland; Postgraduate School of Molecular Medicine, Warsaw, Poland
| | - Jacek Kuznicki
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland
| | - Marta B Wisniewska
- International Institute of Molecular and Cell Biology, Laboratory of Neurodegeneration, Warsaw, Poland; University of Warsaw, Centre of New Technologies, Laboratory of Molecular Neurobiology, Poland.
| |
Collapse
|
12
|
Sittig LJ, Carbonetto P, Engel KA, Krauss KS, Barrios-Camacho CM, Palmer AA. Genetic Background Limits Generalizability of Genotype-Phenotype Relationships. Neuron 2016; 91:1253-1259. [PMID: 27618673 PMCID: PMC5033712 DOI: 10.1016/j.neuron.2016.08.013] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/11/2016] [Accepted: 08/08/2016] [Indexed: 01/16/2023]
Abstract
Genome-wide association studies (GWASs) have identified numerous loci that influence risk for psychiatric diseases. Genetically engineered mice are often used to characterize genes implicated by GWASs. These studies are based on the assumption that observed genotype-phenotype relationships will generalize to humans, implying that the results would at least generalize to other inbred mouse strains. Given current concerns about reproducibility, we sought to directly test this assumption. We produced F1 crosses between male C57BL/6J mice heterozygous for null alleles of Cacna1c and Tcf7l2 and wild-type females from 30 inbred laboratory strains. We found extremely strong interactions with genetic background that sometimes supported diametrically opposing conclusions. These results do not negate the invaluable contributions of mouse genetics to biomedical science, but they do show that genotype-phenotype relationships cannot be reliably inferred by studying a single genetic background, and thus constitute a major challenge to the status quo. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Laura J Sittig
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA; Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Kyle A Engel
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Kathleen S Krauss
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Camila M Barrios-Camacho
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Abraham A Palmer
- Department of Human Genetics, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA; Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Nagalski A, Puelles L, Dabrowski M, Wegierski T, Kuznicki J, Wisniewska MB. Molecular anatomy of the thalamic complex and the underlying transcription factors. Brain Struct Funct 2016; 221:2493-510. [PMID: 25963709 PMCID: PMC4884203 DOI: 10.1007/s00429-015-1052-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 04/27/2015] [Indexed: 01/19/2023]
Abstract
Thalamocortical loops have been implicated in the control of higher-order cognitive functions, but advances in our understanding of the molecular underpinnings of neocortical organization have not been accompanied by similar analyses in the thalamus. Using expression-based correlation maps and the manual mapping of mouse and human datasets available in the Allen Brain Atlas, we identified a few individual regions and several sets of molecularly related nuclei that partially overlap with the classic grouping that is based on topographical localization and thalamocortical connections. These new molecular divisions of the adult thalamic complex are defined by the combinatorial expression of Tcf7l2, Lef1, Gbx2, Prox1, Pou4f1, Esrrg, and Six3 transcription factor genes. Further in silico and experimental analyses provided the evidence that TCF7L2 might be a pan-thalamic specifier. These results provide substantial insights into the "molecular logic" that underlies organization of the thalamic complex.
Collapse
Affiliation(s)
- Andrzej Nagalski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, 00-927, Poland
| | - Luis Puelles
- Department of Human Anatomy, University of Murcia and IMIB, Murcia, 30071, Spain
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Center of Neurobiology, Nencki Institute of Experimental Biology, Warsaw, 02-093, Poland
| | - Tomasz Wegierski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
| | - Marta B Wisniewska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland.
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, 00-927, Poland.
| |
Collapse
|
14
|
Hamamura K, Matsunaga N, Ikeda E, Kondo H, Ikeyama H, Tokushige K, Itcho K, Furuichi Y, Yoshida Y, Matsuda M, Yasuda K, Doi A, Yokota Y, Amamoto T, Aramaki H, Irino Y, Koyanagi S, Ohdo S. Alterations of Hepatic Metabolism in Chronic Kidney Disease via D-box-binding Protein Aggravate the Renal Dysfunction. J Biol Chem 2016; 291:4913-27. [PMID: 26728457 DOI: 10.1074/jbc.m115.696930] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 11/06/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with an increase in serum retinol; however, the underlying mechanisms of this disorder are poorly characterized. Here, we found that the alteration of hepatic metabolism induced the accumulation of serum retinol in 5/6 nephrectomy (5/6Nx) mice. The liver is the major organ responsible for retinol metabolism; accordingly, microarray analysis revealed that the hepatic expression of most CYP genes was changed in 5/6Nx mice. In addition, D-box-binding protein (DBP), which controls the expression of several CYP genes, was significantly decreased in these mice. Cyp3a11 and Cyp26a1, encoding key proteins in retinol metabolism, showed the greatest decrease in expression in 5/6Nx mice, a process mediated by the decreased expression of DBP. Furthermore, an increase of plasma transforming growth factor-β1 (TGF-β1) in 5/6Nx mice led to the decreased expression of the Dbp gene. Consistent with these findings, the alterations of retinol metabolism and renal dysfunction in 5/6Nx mice were ameliorated by administration of an anti-TGF-β1 antibody. We also show that the accumulation of serum retinol induced renal apoptosis in 5/6Nx mice fed a normal diet, whereas renal dysfunction was reduced in mice fed a retinol-free diet. These findings indicate that constitutive Dbp expression plays an important role in mediating hepatic dysfunction under CKD. Thus, the aggravation of renal dysfunction in patients with CKD might be prevented by a recovery of hepatic function, potentially through therapies targeting DBP and retinol.
Collapse
Affiliation(s)
- Kengo Hamamura
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan, the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Naoya Matsunaga
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Eriko Ikeda
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan, the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Hideaki Kondo
- the Center for Sleep Medicine, Saiseikai Nagasaki Hospital, Katafuchi, Nagasaki 850-0003, Japan
| | - Hisako Ikeyama
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kazutaka Tokushige
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kazufumi Itcho
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yoko Furuichi
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yuya Yoshida
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Masaki Matsuda
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kaori Yasuda
- Cell-Innovator Inc., EC Building, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Atsushi Doi
- Cell-Innovator Inc., EC Building, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yoshifumi Yokota
- the Department of Molecular Genetics, School of Medicine, Fukui University, Matsuoka, Fukui 910-1193, Japan
| | - Toshiaki Amamoto
- Neues Corporation, Tenyamachi-cho, Hakata-ku, Fukuoka 812-0025, Japan, and
| | - Hironori Aramaki
- the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan, the Department of Molecular Biology, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Yasuhiro Irino
- the Integrated Center for Mass Spectrometry, Division of Membrane Biology, and Department of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Satoru Koyanagi
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Shigehiro Ohdo
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan,
| |
Collapse
|
15
|
Abstract
INTRODUCTION Electroconvulsive therapy (ECT) is currently regarded as a significant treatment option for intractable psychiatric disorders, such as catatonic schizophrenia or treatment-resistant depression; however, the underlying molecular mechanism for its therapeutic effect remains obscure. METHODS Employing microarray analysis (Human Genome U133 Plus 2.0 Array; Affymetrix, United States) of cDNA derived from the peripheral blood of patients with catatonic schizophrenia (n = 5), we detected a significant change in 145 genes (0.68%) before and after modified ECT (mECT). Moreover, we performed quantitative polymerase chain reaction validation of genes that had previously been suggested to be functionally related to schizophrenia. RESULTS Of 4 genes examined (AKT3, TCF7, PPP3R1, and GADD45B), only TCF7 was increased during the mECT procedure (P = 0.0025). DISCUSSION This study describes the first attempt to uncover the molecular mechanism of mECT using a microarray assay of mRNA derived from peripheral blood, and our results suggest that the TCF family may play a role in the functional mechanism of mECT.
Collapse
|
16
|
Zhou Y, Park SY, Su J, Bailey K, Ottosson-Laakso E, Shcherbina L, Oskolkov N, Zhang E, Thevenin T, Fadista J, Bennet H, Vikman P, Wierup N, Fex M, Rung J, Wollheim C, Nobrega M, Renström E, Groop L, Hansson O. TCF7L2 is a master regulator of insulin production and processing. Hum Mol Genet 2014; 23:6419-31. [PMID: 25015099 PMCID: PMC4240194 DOI: 10.1093/hmg/ddu359] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genome-wide association studies have revealed >60 loci associated with type 2 diabetes (T2D), but the underlying causal variants and functional mechanisms remain largely elusive. Although variants in TCF7L2 confer the strongest risk of T2D among common variants by presumed effects on islet function, the molecular mechanisms are not yet well understood. Using RNA-sequencing, we have identified a TCF7L2-regulated transcriptional network responsible for its effect on insulin secretion in rodent and human pancreatic islets. ISL1 is a primary target of TCF7L2 and regulates proinsulin production and processing via MAFA, PDX1, NKX6.1, PCSK1, PCSK2 and SLC30A8, thereby providing evidence for a coordinated regulation of insulin production and processing. The risk T-allele of rs7903146 was associated with increased TCF7L2 expression, and decreased insulin content and secretion. Using gene expression profiles of 66 human pancreatic islets donors’, we also show that the identified TCF7L2-ISL1 transcriptional network is regulated in a genotype-dependent manner. Taken together, these results demonstrate that not only synthesis of proinsulin is regulated by TCF7L2 but also processing and possibly clearance of proinsulin and insulin. These multiple targets in key pathways may explain why TCF7L2 has emerged as the gene showing one of the strongest associations with T2D.
Collapse
Affiliation(s)
- Yuedan Zhou
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | | | - Jing Su
- European Bioinformatics Institute, Functional Genomics, Hinxton, Cambridge CB10 1SD, UK
| | - Kathleen Bailey
- Department of Human Genetics, University of Chicago, IL 60637, USA
| | | | - Liliya Shcherbina
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Nikolay Oskolkov
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Enming Zhang
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Thomas Thevenin
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - João Fadista
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Hedvig Bennet
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Petter Vikman
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Nils Wierup
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Malin Fex
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Johan Rung
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala 75185, Sweden and
| | - Claes Wollheim
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden, Department of Cell Physiology and Metabolism, Université de Genève, University Medical Centre, 1 rue Michel-Servet, Geneva 4 1211, Switzerland
| | - Marcelo Nobrega
- Department of Human Genetics, University of Chicago, IL 60637, USA
| | - Erik Renström
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Leif Groop
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden
| | - Ola Hansson
- Department of Clinical Sciences, CRC, Lund University, Malmö 20502, Sweden,
| |
Collapse
|
17
|
Nagalski A, Irimia M, Szewczyk L, Ferran JL, Misztal K, Kuznicki J, Wisniewska MB. Postnatal isoform switch and protein localization of LEF1 and TCF7L2 transcription factors in cortical, thalamic, and mesencephalic regions of the adult mouse brain. Brain Struct Funct 2013; 218:1531-49. [PMID: 23152144 PMCID: PMC3825142 DOI: 10.1007/s00429-012-0474-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 10/25/2012] [Indexed: 02/06/2023]
Abstract
β-Catenin signaling, leading to the activation of lymphoid enhancer-binding factor 1/T cell factor (LEF1/TCF) transcription factors, plays a well-established role in transcription regulation during development and tissue homeostasis. In the adult organism, the activity of this pathway has been found in stem cell niches and postmitotic thalamic neurons. Recently, studies show that mutations in components of β-catenin signaling networks have been associated with several psychiatric disorders, indicating the involvement of β-catenin and LEF1/TCF proteins in the proper functioning of the brain. Here, we report a comprehensive analysis of LEF1/TCF protein localization and the expression profile of their isoforms in cortical, thalamic, and midbrain regions in mice. We detected LEF1 and TCF7L2 proteins in neurons of the thalamus and dorsal midbrain, i.e., subcortical regions specialized in the integration of diverse sources of sensory information. These neurons also exhibited nuclear localization of β-catenin, suggesting the involvement of β-catenin/LEF1/TCF7L2 in the regulation of gene expression in these regions. Analysis of alternative splicing and promoter usage identified brain-specific TCF7L2 isoforms and revealed a developmentally coordinated transition in the composition of LEF1 and TCF7L2 isoforms. In the case of TCF7L2, the typical brain isoforms lack the so-called C clamp; in addition, the dominant-negative isoforms are predominant in the embryonic thalamus but disappear postnatally. The present study provides a necessary framework to understand the role of LEF1/TCF factors in thalamic and midbrain development until adulthood and predicts that the regulatory role of these proteins in the adult brain is significantly different from their role in the embryonic brain or other non-neural tissues.
Collapse
Affiliation(s)
- A. Nagalski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| | - M. Irimia
- Banting and Best Department of Medical Research, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - L. Szewczyk
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| | - J. L. Ferran
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, E30071 Spain
| | - K. Misztal
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| | - J. Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - M. B. Wisniewska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| |
Collapse
|
18
|
Wisniewska MB. Physiological role of β-catenin/TCF signaling in neurons of the adult brain. Neurochem Res 2013; 38:1144-55. [PMID: 23377854 PMCID: PMC3653035 DOI: 10.1007/s11064-013-0980-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 12/21/2012] [Accepted: 01/19/2013] [Indexed: 12/21/2022]
Abstract
Wnt/β-catenin pathway, the effectors of which are transcription factors of the LEF1/TCF family, is primarily associated with development. Strikingly, however, some of the genes of the pathway are schizophrenia susceptibility genes, and the proteins that are often mutated in neurodegenerative diseases have the ability to regulate β-catenin levels. If impairment of this pathway indeed leads to these pathologies, then it likely plays a physiological role in the adult brain. This review provides an overview of the current knowledge on this subject. The involvement of β-catenin and LEF1/TCF factors in adult neurogenesis, synaptic plasticity, and the function of thalamic neurons are discussed. The data are still very preliminary and often based on circumstantial or indirect evidence. Further research might help to understand the etiology of the aforementioned pathologies.
Collapse
Affiliation(s)
- Marta B Wisniewska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, ul. Ks. Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
19
|
Savic D, Bell GI, Nobrega MA. An in vivo cis-regulatory screen at the type 2 diabetes associated TCF7L2 locus identifies multiple tissue-specific enhancers. PLoS One 2012; 7:e36501. [PMID: 22590553 PMCID: PMC3349716 DOI: 10.1371/journal.pone.0036501] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/02/2012] [Indexed: 12/21/2022] Open
Abstract
Genome-wide association studies (GWAS) have repeatedly shown an association between non-coding variants in the TCF7L2 locus and risk for type 2 diabetes (T2D), implicating a role for cis-regulatory variation within this locus in disease etiology. Supporting this hypothesis, we previously localized complex regulatory activity to the TCF7L2 T2D-associated interval using an in vivo bacterial artificial chromosome (BAC) enhancer-trapping reporter strategy. To follow-up on this broad initial survey of the TCF7L2 regulatory landscape, we performed a fine-mapping enhancer scan using in vivo mouse transgenic reporter assays. We functionally interrogated approximately 50% of the sequences within the T2D-associated interval, utilizing sequence conservation within this 92-kb interval to determine the regulatory potential of all evolutionary conserved sequences that exhibited conservation to the non-eutherian mammal opossum. Included in this study was a detailed functional interrogation of sequences spanning both protective and risk alleles of single nucleotide polymorphism (SNP) rs7903146, which has exhibited allele-specific enhancer function in pancreatic beta cells. Using these assays, we identified nine segments regulating various aspects of the TCF7L2 expression profile and that constitute nearly 70% of the sequences tested. These results highlight the regulatory complexity of this interval and support the notion that a TCF7L2 cis-regulatory disruption leads to T2D predisposition.
Collapse
Affiliation(s)
- Daniel Savic
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America.
| | | | | |
Collapse
|