1
|
LaMonte OC, Feldman SR. Indoor Tanning Addiction: Biological Mechanisms and Association with Other Disorders. J Cutan Med Surg 2024:12034754241303135. [PMID: 39680097 DOI: 10.1177/12034754241303135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Although many people who use tanning beds are aware of the negative consequences, they continue to indoor tan, possibly due to addictive properties. The purpose of this paper is to review the existing literature on tanning addiction, its potential biological mechanisms, and its association with psychological disorders. A PubMed search was conducted using the terms "Tanning Addiction," "UVR AND B-endorphin," and "tanning dependence AND gene." The articles were organized based on the following categories: endorphins, dopamine and the brain, genetic associations, and associations between tanning addiction and other disorders. Exposure to ultraviolet radiation leads to the synthesis of β-endorphins in the skin, the release of dopamine in the brain, and increased blood flow in brain regions associated with reinforcement and addiction. Genes associated with tanning addiction include PTCHD2 and ANKK1. Several psychiatric disorders are comorbidities of tanning addiction and excessive indoor tanning parallels many of the patterns of substance use disorders. In frequent indoor tanners, greater awareness of tanning's addictive potential and further development of interventions aimed at treating this addictive behaviour may be needed to stop indoor tanning.
Collapse
Affiliation(s)
- Olivia C LaMonte
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
2
|
Li D, Liu Z, Zhang L, Bian X, Wu J, Li L, Chen Y, Luo L, Pan L, Kong L, Xiao Y, Wang J, Zhang X, Wang W, Toma M, Piipponen M, Sommar P, Xu Landén N. The lncRNA SNHG26 drives the inflammatory-to-proliferative state transition of keratinocyte progenitor cells during wound healing. Nat Commun 2024; 15:8637. [PMID: 39366968 PMCID: PMC11452505 DOI: 10.1038/s41467-024-52783-8] [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: 12/26/2023] [Accepted: 09/18/2024] [Indexed: 10/06/2024] Open
Abstract
The cell transition from an inflammatory phase to a subsequent proliferative phase is crucial for wound healing, yet the driving mechanism remains unclear. By profiling lncRNA expression changes during human skin wound healing and screening lncRNA functions, we identify SNHG26 as a pivotal regulator in keratinocyte progenitors underpinning this phase transition. Snhg26-deficient mice exhibit impaired wound repair characterized by delayed re-epithelization accompanied by exacerbated inflammation. Single-cell transcriptome analysis combined with gain-of-function and loss-of-function of SNHG26 in vitro and ex vivo reveals its specific role in facilitating inflammatory-to-proliferative state transition of keratinocyte progenitors. A mechanistic study unravels that SNHG26 interacts with and relocates the transcription factor ILF2 from inflammatory genomic loci, such as JUN, IL6, IL8, and CCL20, to the genomic locus of LAMB3. Collectively, our findings suggest that lncRNAs play cardinal roles in expediting tissue repair and regeneration and may constitute an invaluable reservoir of therapeutic targets in reparative medicine.
Collapse
Affiliation(s)
- Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China.
| | - Zhuang Liu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Letian Zhang
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Xiaowei Bian
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Jianmin Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Institute of Genomic Medicine, Wenzhou Medical University, 325035, Wenzhou, China
| | - Li Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Lihua Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Ling Pan
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Lingzhuo Kong
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Yunting Xiao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Jiating Wang
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Xiya Zhang
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Wang Wang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Maria Toma
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Minna Piipponen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Pehr Sommar
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden.
| |
Collapse
|
3
|
Li M, Na X, Lin F, Liang S, Huang Y, Song J, Xu X, Yang C. DMF-ChIP-seq for Highly Sensitive and Integrated Epigenomic Profiling of Low-Input Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:52047-52058. [PMID: 39303213 DOI: 10.1021/acsami.4c11280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Mapping genome-wide DNA-protein interactions (DPIs) provides insights into the epigenetic landscape of complex biological systems and elucidates the mechanisms of epigenetic regulation in biological progress. However, current technologies in DPI profiling still suffer from high cell demands, low detection sensitivity, and large reagent consumption. To address these problems, we developed DMF-ChIP-seq that builds on digital microfluidic (DMF) technology to profile genome-wide DPIs in a highly efficient, cost-effective, and user-friendly way. The entire workflow including cell pretreatment, antibody recognition, pA-Tn5 tagmentation, fragment enrichment, and PCR amplification is programmatically manipulated on a single chip. Leveraging closed submicroliter reaction volumes and a superhydrophobic interface, DMF-ChIP-seq presented higher sensitivity in peak enrichment than other current methods, with high accuracy (Pearson Correlation Coefficient (PCC) > 0.86) and high repeatability (PCC > 0.92). Furthermore, DMF-ChIP-seq was capable of processing the samples with as few as 8 cells while maintaining a high signal-to-noise ratio. By applying DMF-ChIP-seq, H3K27ac histone modification of early embryonic cells during differentiation was profiled for the investigation of epigenomic landscape dynamics. With the benefits of high efficiency and sensitivity in DPI analysis, the system provides great promise in studying epigenetic regulation during various biological processes.
Collapse
Affiliation(s)
- Mingyin Li
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xing Na
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Fanghe Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shanshan Liang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yuehan Huang
- School of International Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jia Song
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xing Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Technology for Precision Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
4
|
Monasso GS, Hoang TT, Mancano G, Fernández-Barrés S, Dou J, Jaddoe VW, Page CM, Johnson L, Bustamante M, Bakulski KM, Håberg SE, Ueland PM, Battram T, Merid SK, Melén E, Caramaschi D, Küpers LK, Sunyer J, Nystad W, Heil SG, Schmidt RJ, Vrijheid M, Sharp GC, London SJ, Felix JF. A meta-analysis of epigenome-wide association studies on pregnancy vitamin B12 concentrations and offspring DNA methylation. Epigenetics 2023; 18:2202835. [PMID: 37093107 PMCID: PMC10128528 DOI: 10.1080/15592294.2023.2202835] [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: 07/16/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 04/25/2023] Open
Abstract
Circulating vitamin B12 concentrations during pregnancy are associated with offspring health. Foetal DNA methylation changes could underlie these associations. Within the Pregnancy And Childhood Epigenetics Consortium, we meta-analysed epigenome-wide associations of circulating vitamin B12 concentrations in mothers during pregnancy (n = 2,420) or cord blood (n = 1,029), with cord blood DNA methylation. Maternal and newborn vitamin B12 concentrations were associated with DNA methylation at 109 and 7 CpGs, respectively (False Discovery Rate P-value <0.05). Persistent associations with DNA methylation in the peripheral blood of up to 482 children aged 4-10 y were observed for 40.7% of CpGs associated with maternal vitamin B12 and 57.1% of CpGs associated with newborn vitamin B12. Of the CpGs identified in the maternal meta-analyses, 4.6% were associated with either birth weight or gestational age in a previous work. For the newborn meta-analysis, this was the case for 14.3% of the identified CpGs. Also, of the CpGs identified in the newborn meta-analysis, 14.3% and 28.6%, respectively, were associated with childhood cognitive skills and nonverbal IQ. Of the 109 CpGs associated with maternal vitamin B12, 18.3% were associated with nearby gene expression. In this study, we showed that maternal and newborn vitamin B12 concentrations are associated with DNA methylation at multiple CpGs in offspring blood (PFDR<0.05). Whether this differential DNA methylation underlies associations of vitamin B12 concentrations with child health outcomes, such as birth weight, gestational age, and childhood cognition, should be further examined in future studies.
Collapse
Affiliation(s)
- Giulietta S. Monasso
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Thanh T. Hoang
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Giulia Mancano
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School Population Health Sciences, University of Bristol, Bristol, UK
| | - Sílvia Fernández-Barrés
- ISGlobal, Bacelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), madrid,Barcelona, Spain
| | - John Dou
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, USA
| | - Vincent W.V. Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Christian M. Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Laura Johnson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Centre for Exercise, Nutrition and Health Sciences, School for Policy Studies, University of Bristol, Bristol, UK
| | - Mariona Bustamante
- ISGlobal, Bacelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), madrid,Barcelona, Spain
| | - Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, USA
| | - Siri E. Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Thomas Battram
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Simon K. Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Sciences and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Sciences and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Sachs’ Children’s Hospital, South General Hospital, Stockholm, Sweden
| | - Doretta Caramaschi
- College of Life and Environmental Sciences, Department of Psychology, University of Exeter, Exeter, UK
| | - Leanne K. Küpers
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Jordi Sunyer
- ISGlobal, Bacelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), madrid,Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Wenche Nystad
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Sandra G. Heil
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rebecca J. Schmidt
- Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, USA
- The UC Davis MIND Institute, School of Medicine, University of California Davis, Sacramento, USA
| | - Martine Vrijheid
- ISGlobal, Bacelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), madrid,Barcelona, Spain
| | - Gemma C. Sharp
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School Population Health Sciences, University of Bristol, Bristol, UK
| | - Stephanie J. London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Janine F. Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Heimke M, Richter F, Heinze T, Kunke M, Wedel T, Böttner M, Egberts JH, Lucius R, Cossais F. Localization Pattern of Dispatched Homolog 2 (DISP2) in the Central and Enteric Nervous System. J Mol Neurosci 2023; 73:539-548. [PMID: 37369878 PMCID: PMC10517031 DOI: 10.1007/s12031-023-02129-8] [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: 03/07/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
Dispatched homolog (DISP) proteins have been implicated in the regulation of hedgehog signaling during embryologic development. Although DISP2 has recently been associated with neuronal development and control of cognitive functions, its localization pattern in the mammalian central and peripheral nervous system has not yet been investigated. In this study, the Disp2 expression profile was assessed in human tissues from publicly available transcriptomic datasets. The DISP2 localization pattern was further characterized in the human and rat central nervous system (CNS), as well as within the colonic enteric nervous system (ENS) using dual-label immunohistochemistry. Colocalization of DISP2 with neuronal and glial markers was additionally analyzed in murine primary ENS culture. At transcriptomic level, DISP2 expression was predominant in neuronal cell types of the CNS and ENS. DISP2 immunoreactivity was mainly located within PGP9.5-positive neurons rather than in S100-positive glial cells throughout the nervous system. Investigation of human and rat brain tissues, colonic specimens, and murine ENS primary cultures revealed that DISP2 was located in neuronal cell somata, as well as along neuronal processes both in the human and murine CNS and ENS. Our results indicate that DISP2 is prominently localized within neuronal cells of the CNS and ENS and support putative functions of DISP2 in these tissues.
Collapse
Affiliation(s)
- Marvin Heimke
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Florian Richter
- Department of General, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Tillmann Heinze
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Madlen Kunke
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Thilo Wedel
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Martina Böttner
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | | | - Ralph Lucius
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - François Cossais
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany.
| |
Collapse
|
6
|
Oliveira MDS, Cassiano LMG, Pioline J, de Carvalho KRA, Salim ACDM, Alves PA, Fernandes GDR, Machado ADMV, Coimbra RS. Organotypic hippocampal culture model reveals differential responses to highly similar Zika virus isolates. J Neuroinflammation 2023; 20:140. [PMID: 37301965 DOI: 10.1186/s12974-023-02826-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
INTRODUCTION Zika virus (ZIKV) caused an outbreak in Brazil, in 2015, being associated to microcephaly. ZIKV has a strong neurotropism leading to death of infected cells in different brain regions, including the hippocampus, a major site for neurogenesis. The neuronal populations of the brain are affected differently by ZIKV from Asian and African ancestral lineages. However, it remains to be investigated whether subtle variations in the ZIKV genome can impact hippocampus infection dynamics and host response. OBJECTIVE This study evaluated how two Brazilian ZIKV isolates, PE243 and SPH2015, that differ in two specific missense amino acid substitutions, one in the NS1 protein and the other in the NS4A protein, affect the hippocampal phenotype and transcriptome. METHODS Organotypic hippocampal cultures (OHC) from infant Wistar rats were infected with PE243 or SPH2015 and analyzed in time series using immunofluorescence, confocal microscopy, RNA-Seq and RT-qPCR. RESULTS Unique patterns of infection and changes in neuronal density in the OHC were observed for PE243 and SPH2015 between 8 and 48 h post infection (p.i.). Phenotypic analysis of microglia indicated that SPH2015 has a greater capacity for immune evasion. Transcriptome analysis of OHC at 16 h p.i. disclosed 32 and 113 differentially expressed genes (DEGs) in response to infection with PE243 and SPH2015, respectively. Functional enrichment analysis suggested that infection with SPH2015 activates mostly astrocytes rather than microglia. PE243 downregulated biological process of proliferation of brain cells and upregulated those associated with neuron death, while SPH2015 downregulated processes related to neuronal development. Both isolates downregulated cognitive and behavioral development processes. Ten genes were similarly regulated by both isolates. They are putative biomarkers of early hippocampus response to ZIKV infection. At 5, 7, and 10 days p.i., neuronal density of infected OHC remained below controls, and mature neurons of infected OHC showed an increase in the epigenetic mark H3K4me3, which is associated to a transcriptionally active state. This feature is more prominent in response to SPH2015. CONCLUSION Subtle genetic diversity of the ZIKV affects the dynamics of viral dissemination in the hippocampus and host response in the early stages of infection, which may lead to different long-term effects in neuronal population.
Collapse
Affiliation(s)
| | - Larissa Marcely Gomes Cassiano
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jeanne Pioline
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil
- Aix-Marseille University, Marseille, France
| | | | - Anna Christina de Matos Salim
- Plataforma de Sequenciamento NGS (Next Generation Sequencing), Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil
| | - Pedro Augusto Alves
- Imunologia de Doenças Virais, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil
| | | | | | - Roney Santos Coimbra
- Neurogenômica, Imunopatologia, Instituto René Rachou, Fiocruz, Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Borda V, da Silva Francisco Junior R, Carvalho JB, Morais GL, Duque Rossi Á, Pezzuto P, Azevedo GS, Schamber-Reis BL, Portari EA, Melo A, Moreira MEL, Guida LC, Cunha DP, Gomes L, Vasconcelos ZFM, Faucz FR, Tanuri A, Stratakis CA, Aguiar RS, Cardoso CC, de Vasconcelos ATR. Whole-exome sequencing reveals insights into genetic susceptibility to Congenital Zika Syndrome. PLoS Negl Trop Dis 2021; 15:e0009507. [PMID: 34125832 PMCID: PMC8224898 DOI: 10.1371/journal.pntd.0009507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 06/24/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Congenital Zika Syndrome (CZS) is a critical illness with a wide range of severity caused by Zika virus (ZIKV) infection during pregnancy. Life-threatening neurodevelopmental dysfunctions are among the most common phenotypes observed in affected newborns. Risk factors that contribute to susceptibility and response to ZIKV infection may be related to the virus itself, the environment, and maternal genetic background. Nevertheless, the newborn’s genetic contribution to the critical illness is still not elucidated. Here, we aimed to identify possible genetic variants as well as relevant biological pathways that might be associated with CZS phenotypes. For this purpose, we performed a whole-exome sequencing in 40 children born to women with confirmed exposure to ZIKV during pregnancy. We investigated the occurrence of rare harmful single-nucleotide variants (SNVs) possibly associated with inborn errors in genes ontologically related to CZS phenotypes. Moreover, an exome-wide association analysis was also performed using a case-control design (29 CZS cases and 11 controls), for both common and rare variants. Five out of the 29 CZS patients harbored known pathogenic variants likely to contribute to mild to severe manifestations observed. Approximately, 30% of affected individuals carried at least one pathogenic or likely pathogenic SNV in genes candidates to play a role in CZS. Our common variant association analysis detected a suggestive protective effect of the rs2076469 in DISP3 gene (p-value: 1.39 x 10−5). The IL12RB2 gene (p-value: 2.18x10-11) also showed an unusual distribution of nonsynonymous rare SNVs in control samples. Finally, genes harboring harmful variants are involved in processes related to CZS phenotypes such as neurological development and immunity. Therefore, both rare and common variations may be likely to contribute as the underlying genetic cause of CZS susceptibility. The variations and pathways identified in this study may also have implications for the development of therapeutic strategies in the future. Since the beginning of Zika virus outbreak in Brazil, five years ago, we still don’t understand the genetic factors associated with the small number of babies born with Congenital Zika Syndrome (CZS). Here, we focused on the host genetic susceptibility by studying the whole-exome of the CZS affected (n = 29) and healthy (n = 11) neonates, both born to ZIKV infected women from Brazil. We applied two strategies: 1) Determine whether cases individuals have pathogenic or harmful variants that explain the CZS outcomes (i.e. microcephaly) independently of ZIKV infection or not, 2) Exploring the common and rare variants association with CZS. We found that common and rare variants in genes like DISP3 and IL12RB2 could explain some level of the susceptibility to CZS. Moreover, by considering these and other candidate genes, we observed an over-representation of Gene Ontology terms related to neurological system, metabolism and microtubule-cytoskeleton organization.
Collapse
Affiliation(s)
- Victor Borda
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica LNCC/MCTIC Petrópolis, Brazil
| | | | - Joseane B. Carvalho
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica LNCC/MCTIC Petrópolis, Brazil
| | - Guilherme L. Morais
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica LNCC/MCTIC Petrópolis, Brazil
| | - Átila Duque Rossi
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Pezzuto
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Bruno L. Schamber-Reis
- Faculdade de Ciências Médicas de Campina Grande, Núcleo de Genética Médica, Centro Universitário UniFacisa, Campina Grande, Brazil
| | | | - Adriana Melo
- Instituto de Pesquisa Professor Amorim Neto, Campina Grande Brazil
- Faculdade de Ciências Médicas de Campina Grande, Núcleo de Genética Médica, Centro Universitário UniFacisa, Campina Grande, Brazil
| | | | | | | | - Leonardo Gomes
- Instituto Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | | | - Fabio R. Faucz
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Renato S. Aguiar
- Departamento de Genética, Ecologia e Evolução Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- * E-mail: (RSA); (CCC); (ATRV)
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (RSA); (CCC); (ATRV)
| | - Ana Tereza Ribeiro de Vasconcelos
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica LNCC/MCTIC Petrópolis, Brazil
- * E-mail: (RSA); (CCC); (ATRV)
| |
Collapse
|
8
|
Wang W, Li W, Jiang W, Lin H, Wu Y, Wen Y, Xu C, Tian X, Li S, Tan Q, Zhang D. Genome-wide DNA methylation analysis of cognitive function in middle and old-aged Chinese monozygotic twins. J Psychiatr Res 2021; 136:571-580. [PMID: 33131831 DOI: 10.1016/j.jpsychires.2020.10.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 08/13/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022]
Abstract
Cognitive ability plays an important role in mental and physical well-beings in the increasingly ageing populations. Here, based on a sample of 30 cognitive function-discordant monozygotic twin pairs, we aimed to detect specific epigenetic variants potentially related to cognitive function by conducting an epigenome-wide association study (EWAS). Association between methylation level of single CpG site with cognitive function score was tested by linear mixed effect model. Functions of cis-regulatory regions and ontology enrichments were predicted by Genomic Regions Enrichment of Annotations Tool (GREAT). Differentially methylated regions (DMRs) were detected by comb-p python library. A list of 28 CpG sites were identified to reach the level of P < 1 × 10-4, and the strongest association (cor = 0.138, P = 2.549 × 10-6) was detected for DNA CpG site (Chr17: 40,700,490 bp) located at HSD17B1P1. The identified 14,065 genomic CpG sites (P < 0.05) were mapped to 2646 genes, especially HSD17B1P1, CUL4A, INTS8, GFI1B, ZNF467, CDH15, and PSMA1. GREAT ontology enrichments mainly highlighted nicotine pharmacodynamics pathway, GABA-B receptor II/nicotinic acetylcholine receptor/hedgehog/endothelin/Wnt signaling pathways, Parkinson disease, Huntington disease, glycolysis, neuronal system, and toll-like receptor binding. We detected 15 DMRs located at/near 16 genes, especially LINC01551, LINC02282, and FAM32A. And 32 cognitive function-associated differentially methylated genes could be replicated, such as SHANK2, ABCA2, PRDM16, NCOR2, and INPP5A. Our EWAS in monozygotic twins identify specific epigenetic variations which are significantly involved in functional genes, biological function and pathways that mediate cognitive function. The findings provide clues to further identify new diagnostic biomarkers and therapeutic targets for cognitive dysfunction.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, 266021, Shandong Province, China.
| | - Weilong Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, DK-5000, Odense C, Denmark.
| | - Wenjie Jiang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, 266021, Shandong Province, China.
| | - Haijun Lin
- Biomarker Technologies orporation, Beijing, 100000, China.
| | - Yili Wu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, 266021, Shandong Province, China.
| | - Yanhua Wen
- Biomarker Technologies orporation, Beijing, 100000, China.
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, 266033, Shandong Province, China; Qingdao Institute of Preventive Medicine, Qingdao, 266033, Shandong Province, China.
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, 266033, Shandong Province, China; Qingdao Institute of Preventive Medicine, Qingdao, 266033, Shandong Province, China.
| | - Shuxia Li
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, DK-5000, Odense C, Denmark.
| | - Qihua Tan
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, DK-5000, Odense C, Denmark; Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, DK-5000, Odense C, Denmark.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, 266021, Shandong Province, China.
| |
Collapse
|
9
|
Profiling NSD3-dependent neural crest gene expression reveals known and novel candidate regulatory factors. Dev Biol 2021; 475:118-130. [PMID: 33705737 DOI: 10.1016/j.ydbio.2021.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/17/2021] [Accepted: 02/21/2021] [Indexed: 12/17/2022]
Abstract
The lysine methyltransferase NSD3 is required for the expression of key neural crest transcription factors and the migration of neural crest cells. Nevertheless, a complete view of the genes dependent upon NSD3 for expression and the developmental processes impacted by NSD3 in the neural crest was lacking. We used RNA sequencing (RNA-seq) to profile transcripts differentially expressed after NSD3 knockdown in chick premigratory neural crest cells, identifying 674 genes. Gene Ontology and gene set enrichment analyses further support a requirement for NSD3 during neural crest development and show that NSD3 knockdown also upregulates ribosome biogenesis. To validate our results, we selected three genes not previously associated with neural crest development, Astrotactin 1 (Astn1), Dispatched 3 (Disp3), and Tropomyosin 1 (Tpm1). Using whole mount in situ hybridization, we show that premigratory neural crest cells express these genes and that NSD3 knockdown downregulates (Astn1 and Disp3) and upregulates (Tpm1) their expression, consistent with RNA-seq results. Altogether, this study identifies novel putative regulators of neural crest development and provides insight into the transcriptional consequences of NSD3 in the neural crest, with implications for cancer.
Collapse
|
10
|
Macron C, Lane L, Núñez Galindo A, Dayon L. Deep Dive on the Proteome of Human Cerebrospinal Fluid: A Valuable Data Resource for Biomarker Discovery and Missing Protein Identification. J Proteome Res 2018; 17:4113-4126. [PMID: 30124047 DOI: 10.1021/acs.jproteome.8b00300] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cerebrospinal fluid (CSF) is a body fluid of choice for biomarker studies of brain disorders but remains relatively under-studied compared with other biological fluids such as plasma, partly due to the more invasive means of its sample collection. The present study establishes an in-depth CSF proteome through the analysis of a unique CSF sample from a pool of donors. After immunoaffinity depletion, the CSF sample was fractionated using off-gel electrophoresis and analyzed with liquid chromatography tandem mass spectrometry (MS) using the latest generation of hybrid Orbitrap mass spectrometers. The shotgun proteomic analysis allowed the identification of 20 689 peptides mapping on 3379 proteins. To the best of our knowledge, the obtained data set constitutes the largest CSF proteome published so far. Among the CSF proteins identified, 34% correspond to genes whose transcripts are highly expressed in brain according to the Human Protein Atlas. The principal Alzheimer's disease biomarkers (e.g., tau protein, amyloid-β, apolipoprotein E, and neurogranin) were detected. Importantly, our data set significantly contributes to the Chromosome-centric Human Proteome Project (C-HPP), and 12 proteins considered as missing are proposed for validation in accordance with the HPP guidelines. Of these 12 proteins, 8 proteins are based on 2 to 6 uniquely mapping peptides from this CSF analysis, and 4 match a new peptide with a "stranded" single peptide in PeptideAtlas from previous CSF studies. The MS proteomic data are available to the ProteomeXchange Consortium ( http://www.proteomexchange.org/ ) with the data set identifier PXD009646.
Collapse
Affiliation(s)
- Charlotte Macron
- Proteomics , Nestlé Institute of Health Sciences , 1015 Lausanne , Switzerland
| | - Lydie Lane
- CALIPHO Group , SIB-Swiss Institute of Bioinformatics , CMU, rue Michel-Servet 1 , 1211 Geneva 4 , Switzerland.,Department of Microbiology and Molecular Medicine, Faculty of Medicine , University of Geneva , rue Michel-Servet 1 , 1211 Geneva 4 , Switzerland
| | | | - Loïc Dayon
- Proteomics , Nestlé Institute of Health Sciences , 1015 Lausanne , Switzerland
| |
Collapse
|
11
|
Detecting Rare Mutations with Heterogeneous Effects Using a Family-Based Genetic Random Field Method. Genetics 2018; 210:463-476. [PMID: 30104420 DOI: 10.1534/genetics.118.301266] [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] [Received: 04/04/2018] [Accepted: 07/29/2018] [Indexed: 01/19/2023] Open
Abstract
The genetic etiology of many complex diseases is highly heterogeneous. A complex disease can be caused by multiple mutations within the same gene or mutations in multiple genes at various genomic loci. Although these disease-susceptibility mutations can be collectively common in the population, they are often individually rare or even private to certain families. Family-based studies are powerful for detecting rare variants enriched in families, which is an important feature for sequencing studies due to the heterogeneous nature of rare variants. In addition, family designs can provide robust protection against population stratification. Nevertheless, statistical methods for analyzing family-based sequencing data are underdeveloped, especially those accounting for heterogeneous etiology of complex diseases. In this article, we introduce a random field framework for detecting gene-phenotype associations in family-based sequencing studies, referred to as family-based genetic random field (FGRF). Similar to existing family-based association tests, FGRF could utilize within-family and between-family information separately or jointly to test an association. We demonstrate that FGRF has comparable statistical power with existing methods when there is no genetic heterogeneity, but can improve statistical power when there is genetic heterogeneity across families. The proposed method also shares the same advantages with the conventional family-based association tests (e.g., being robust to population stratification). Finally, we applied the proposed method to a sequencing data from the Minnesota Twin Family Study, and revealed several genes, including SAMD14, potentially associated with alcohol dependence.
Collapse
|
12
|
Ptchd1 deficiency induces excitatory synaptic and cognitive dysfunctions in mouse. Mol Psychiatry 2018; 23:1356-1367. [PMID: 28416808 PMCID: PMC5984103 DOI: 10.1038/mp.2017.39] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/18/2017] [Accepted: 01/25/2017] [Indexed: 12/12/2022]
Abstract
Synapse development and neuronal activity represent fundamental processes for the establishment of cognitive function. Structural organization as well as signalling pathways from receptor stimulation to gene expression regulation are mediated by synaptic activity and misregulated in neurodevelopmental disorders such as autism spectrum disorder (ASD) and intellectual disability (ID). Deleterious mutations in the PTCHD1 (Patched domain containing 1) gene have been described in male patients with X-linked ID and/or ASD. The structure of PTCHD1 protein is similar to the Patched (PTCH1) receptor; however, the cellular mechanisms and pathways associated with PTCHD1 in the developing brain are poorly determined. Here we show that PTCHD1 displays a C-terminal PDZ-binding motif that binds to the postsynaptic proteins PSD95 and SAP102. We also report that PTCHD1 is unable to rescue the canonical sonic hedgehog (SHH) pathway in cells depleted of PTCH1, suggesting that both proteins are involved in distinct cellular signalling pathways. We find that Ptchd1 deficiency in male mice (Ptchd1-/y) induces global changes in synaptic gene expression, affects the expression of the immediate-early expression genes Egr1 and Npas4 and finally impairs excitatory synaptic structure and neuronal excitatory activity in the hippocampus, leading to cognitive dysfunction, motor disabilities and hyperactivity. Thus our results support that PTCHD1 deficiency induces a neurodevelopmental disorder causing excitatory synaptic dysfunction.
Collapse
|
13
|
Lyu J, Song Z, Chen J, Shepard MJ, Song H, Ren G, Li Z, Guo W, Zhuang Z, Shi Y. Whole-exome sequencing of oral mucosal melanoma reveals mutational profile and therapeutic targets. J Pathol 2018; 244:358-366. [PMID: 29230811 DOI: 10.1002/path.5017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/30/2017] [Accepted: 12/05/2017] [Indexed: 12/16/2022]
Abstract
Oral mucosal melanoma (OMM) is a rare and aggressive subtype of melanoma with little known about its pathogenesis or carcinogenesis. We therefore performed whole-exome sequencing (WES) on 19 matched OMM tumor/normal pairs in order to gain insight into potential genetic drivers of tumor formation. For the first time, we describe the comprehensive mutational profile of OMM. Our data suggest that the genetic background of OMM differs from those of other melanoma subtypes. We identified recurrent mutations involving KIT, POLE, PTPRD, PTCHD2, and DMXL2. Notably, copy number analysis revealed recurrently amplified regions of 12q14 (57.9%, containing CDK4) and 5p15 (47.4%, containing TERT). CNV analysis in a separate cohort of 15 samples validated the frequent CNV in CDK4 and TERT. We also observed that the melanocyte development and pigmentation signaling pathway is frequently altered in OMM. Furthermore, our data suggest several altered genes that may be amenable for targeted therapy. We identified one patient with metastatic OMM in our cohort who was identified to harbor a targetable KIT mutation using our WES results. This patient was able to achieve complete remission following implementation of KIT-targeted therapy. These findings provide further insight into the genetic underpinnings of OMM development and suggest that patients with OMM may benefit from WES analysis to identify potential targetable genetic mutations. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jiong Lyu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China.,Department of Stomatology, First Affiliated Hospital of Zhejiang University, Hangzhou, PR China
| | - Zhijian Song
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jianhua Chen
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, PR China
| | - Matthew J Shepard
- Surgical Neurology Branch, National Institute of Neurologic Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA.,Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Hao Song
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Guoxin Ren
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Zhiqiang Li
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, PR China
| | - Wei Guo
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurologic Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yongyong Shi
- Bio-X Institutes, Ministry of Education Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, PR China
| |
Collapse
|
14
|
Modulated DISP3/PTCHD2 expression influences neural stem cell fate decisions. Sci Rep 2017; 7:41597. [PMID: 28134287 PMCID: PMC5278513 DOI: 10.1038/srep41597] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022] Open
Abstract
Neural stem cells (NSCs) are defined by their dual ability to self-renew through mitotic cell division or differentiate into the varied neural cell types of the CNS. DISP3/PTCHD2 is a sterol-sensing domain-containing protein, highly expressed in neural tissues, whose expression is regulated by thyroid hormone. In the present study, we used a mouse NSC line to investigate what effect DISP3 may have on the self-renewal and/or differentiation potential of the cells. We demonstrated that NSC differentiation triggered significant reduction in DISP3 expression in the resulting astrocytes, neurons and oligodendrocytes. Moreover, when DISP3 expression was disrupted, the NSC "stemness" was suppressed, leading to a larger population of cells undergoing spontaneous neuronal differentiation. Conversely, overexpression of DISP3 resulted in increased NSC proliferation. When NSCs were cultured under differentiation conditions, we observed that the lack of DISP3 augmented the number of NSCs differentiating into each of the neural cell lineages and that neuronal morphology was altered. In contrast, DISP3 overexpression resulted in impaired cell differentiation. Taken together, our findings imply that DISP3 may help dictate the NSC cell fate to either undergo self-renewal or switch to the terminal differentiation cell program.
Collapse
|