1
|
Ali M, Huarte OU, Heurtaux T, Garcia P, Rodriguez BP, Grzyb K, Halder R, Skupin A, Buttini M, Glaab E. Single-Cell Transcriptional Profiling and Gene Regulatory Network Modeling in Tg2576 Mice Reveal Gender-Dependent Molecular Features Preceding Alzheimer-Like Pathologies. Mol Neurobiol 2024; 61:541-566. [PMID: 35980567 PMCID: PMC10861719 DOI: 10.1007/s12035-022-02985-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 07/29/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) onset and progression is influenced by a complex interplay of several environmental and genetic factors, one of them gender. Pronounced gender differences have been observed both in the relative risk of developing AD and in clinical disease manifestations. A molecular level understanding of these gender disparities is still missing, but could provide important clues on cellular mechanisms modulating the disease and reveal new targets for gender-oriented disease-modifying precision therapies. We therefore present here a comprehensive single-cell analysis of disease-associated molecular gender differences in transcriptomics data from the neocortex, one of the brain regions most susceptible to AD, in one of the most widely used AD mouse models, the Tg2576 model. Cortical areas are also most commonly used in studies of post-mortem AD brains. To identify disease-linked molecular processes that occur before the onset of detectable neuropathology, we focused our analyses on an age with no detectable plaques and microgliosis. Cell-type specific alterations were investigated at the level of individual genes, pathways, and gene regulatory networks. The number of differentially expressed genes (DEGs) was not large enough to build context-specific gene regulatory networks for each individual cell type, and thus, we focused on the study of cell types with dominant changes and included analyses of changes across the combination of cell types. We observed significant disease-associated gender differences in cellular processes related to synapse organization and reactive oxygen species metabolism, and identified a limited set of transcription factors, including Egr1 and Klf6, as key regulators of many of the disease-associated and gender-dependent gene expression changes in the model. Overall, our analyses revealed significant cell-type specific gene expression changes in individual genes, pathways and sub-networks, including gender-specific and gender-dimorphic changes in both upstream transcription factors and their downstream targets, in the Tg2576 AD model before the onset of overt disease. This opens a window into molecular events that could determine gender-susceptibility to AD, and uncovers tractable target candidates for potential gender-specific precision medicine for AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, 6200, Maastricht, the Netherlands
| | - Oihane Uriarte Huarte
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L‑4362, Esch-Sur-Alzette, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Beatriz Pardo Rodriguez
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- University of the Basque Country, Cell Biology and Histology Department, 48940, Leioa, Vizcaya, Basque Country, Spain
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Physics and Materials Science, University of Luxembourg, 162a av. de la Faïencerie, 1511, Luxembourg, Luxembourg
- Department of Neuroscience, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
2
|
Wong D, Auguste G, Cardenas CLL, Turner AW, Chen Y, Song Y, Ma L, Perry RN, Aherrahrou R, Kuppusamy M, Yang C, Mosquera JV, Dube CJ, Khan MD, Palmore M, Kalra JK, Kavousi M, Peyser PA, Matic L, Hedin U, Manichaikul A, Sonkusare SK, Civelek M, Kovacic JC, Björkegren JL, Malhotra R, Miller CL. FHL5 Controls Vascular Disease-Associated Gene Programs in Smooth Muscle Cells. Circ Res 2023; 132:1144-1161. [PMID: 37017084 PMCID: PMC10147587 DOI: 10.1161/circresaha.122.321692] [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: 07/18/2022] [Accepted: 03/21/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Genome-wide association studies have identified hundreds of loci associated with common vascular diseases, such as coronary artery disease, myocardial infarction, and hypertension. However, the lack of mechanistic insights for many GWAS loci limits their translation into the clinic. Among these loci with unknown functions is UFL1-four-and-a-half LIM (LIN-11, Isl-1, MEC-3) domain 5 (FHL5; chr6q16.1), which reached genome-wide significance in a recent coronary artery disease/ myocardial infarction GWAS meta-analysis. UFL1-FHL5 is also associated with several vascular diseases, consistent with the widespread pleiotropy observed for GWAS loci. METHODS We apply a multimodal approach leveraging statistical fine-mapping, epigenomic profiling, and ex vivo analysis of human coronary artery tissues to implicate FHL5 as the top candidate causal gene. We unravel the molecular mechanisms of the cross-phenotype genetic associations through in vitro functional analyses and epigenomic profiling experiments in coronary artery smooth muscle cells. RESULTS We prioritized FHL5 as the top candidate causal gene at the UFL1-FHL5 locus through expression quantitative trait locus colocalization methods. FHL5 gene expression was enriched in the smooth muscle cells and pericyte population in human artery tissues with coexpression network analyses supporting a functional role in regulating smooth muscle cell contraction. Unexpectedly, under procalcifying conditions, FHL5 overexpression promoted vascular calcification and dysregulated processes related to extracellular matrix organization and calcium handling. Lastly, by mapping FHL5 binding sites and inferring FHL5 target gene function using artery tissue gene regulatory network analyses, we highlight regulatory interactions between FHL5 and downstream coronary artery disease/myocardial infarction loci, such as FOXL1 and FN1 that have roles in vascular remodeling. CONCLUSIONS Taken together, these studies provide mechanistic insights into the pleiotropic genetic associations of UFL1-FHL5. We show that FHL5 mediates vascular disease risk through transcriptional regulation of downstream vascular remodeling gene programs. These transacting mechanisms may explain a portion of the heritable risk for complex vascular diseases.
Collapse
Affiliation(s)
- Doris Wong
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Gaëlle Auguste
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Christian L. Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Yixuan Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Yipei Song
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - R. Noah Perry
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Redouane Aherrahrou
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Chaojie Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Jose Verdezoto Mosquera
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Collin J. Dube
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Mohammad Daud Khan
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Meredith Palmore
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Jaspreet K. Kalra
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, The Netherlands
| | | | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ani Manichaikul
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney, Australia
| | - Johan L.M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, USA
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Rajeev Malhotra
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clint L. Miller
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Faria L, Canato S, Jesus TT, Gonçalves M, Guerreiro PS, Lopes CS, Meireles I, Morais-de-Sá E, Paredes J, Janody F. Activation of an actin signaling pathway in pre-malignant mammary epithelial cells by P-cadherin is essential for transformation. Dis Model Mech 2023; 16:dmm049652. [PMID: 36808468 PMCID: PMC9983776 DOI: 10.1242/dmm.049652] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.
Collapse
Affiliation(s)
- Lídia Faria
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Master Programme in Oncology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Sara Canato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Physiology and Cancer Program, Champalimaud Foundation, Avenida de Brasília, 1400-038 Lisboa, Portugal
| | - Tito T. Jesus
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Margarida Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Patrícia S. Guerreiro
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Vector B2B - Drug Developing - Associação Para Investigação em Biotecnologia, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla S. Lopes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Isabel Meireles
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- FMUP, Medical Faculty of University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Florence Janody
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780-156 Oeiras, Portugal
| |
Collapse
|
4
|
Tabuchi A, Ihara D. SRF in Neurochemistry: Overview of Recent Advances in Research on the Nervous System. Neurochem Res 2022; 47:2545-2557. [PMID: 35668335 DOI: 10.1007/s11064-022-03632-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/20/2022] [Accepted: 05/07/2022] [Indexed: 10/18/2022]
Abstract
Serum response factor (SRF) is a representative transcription factor that plays crucial roles in various biological phenomena by regulating immediate early genes (IEGs) and genes related to cell morphology and motility, among others. Over the years, the signal transduction pathways activating SRF have been clarified and SRF-target genes have been identified. In this overview, we initially briefly summarize the basic biology of SRF and its cofactors, ternary complex factor (TCF) and megakaryoblastic leukemia (MKL)/myocardin-related transcription factor (MRTF). Progress in the generation of nervous system-specific knockout (KO) or genetically modified mice as well as genetic analyses over the last few decades has not only identified novel SRF-target genes but also highlighted the neurochemical importance of SRF and its cofactors. Therefore, here we next present the phenotypes of mice with nervous system-specific KO of SRF or its cofactors by depicting recent findings associated with brain development, plasticity, epilepsy, stress response, and drug addiction, all of which result from function or dysfunction of the SRF axis. Last, we develop a hypothesis regarding the possible involvement of SRF and its cofactors in human neurological disorders including neurodegenerative, psychiatric, and neurodevelopmental diseases. This overview should deepen our understanding, highlight promising future directions for developing novel therapeutic strategies, and lead to illumination of the mechanisms underlying higher brain functions based on neuronal structure and function.
Collapse
Affiliation(s)
- Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
5
|
Gao P, Lyu Q, Ghanam AR, Lazzarotto CR, Newby GA, Zhang W, Choi M, Slivano OJ, Holden K, Walker JA, Kadina AP, Munroe RJ, Abratte CM, Schimenti JC, Liu DR, Tsai SQ, Long X, Miano JM. Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol 2021; 22:83. [PMID: 33722289 PMCID: PMC7962346 DOI: 10.1186/s13059-021-02304-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Most single nucleotide variants (SNVs) occur in noncoding sequence where millions of transcription factor binding sites (TFBS) reside. Here, a comparative analysis of CRISPR-mediated homology-directed repair (HDR) versus the recently reported prime editing 2 (PE2) system was carried out in mice over a TFBS called a CArG box in the Tspan2 promoter. RESULTS Quantitative RT-PCR showed loss of Tspan2 mRNA in aorta and bladder, but not heart or brain, of mice homozygous for an HDR-mediated three base pair substitution in the Tspan2 CArG box. Using the same protospacer, mice homozygous for a PE2-mediated single-base substitution in the Tspan2 CArG box displayed similar cell-specific loss of Tspan2 mRNA; expression of an overlapping long noncoding RNA was also nearly abolished in aorta and bladder. Immuno-RNA fluorescence in situ hybridization validated loss of Tspan2 in vascular smooth muscle cells of HDR and PE2 CArG box mutant mice. Targeted sequencing demonstrated variable frequencies of on-target editing in all PE2 and HDR founders. However, whereas no on-target indels were detected in any of the PE2 founders, all HDR founders showed varying levels of on-target indels. Off-target analysis by targeted sequencing revealed mutations in many HDR founders, but none in PE2 founders. CONCLUSIONS PE2 directs high-fidelity editing of a single base in a TFBS leading to cell-specific loss in expression of an mRNA/long noncoding RNA gene pair. The PE2 platform expands the genome editing toolbox for modeling and correcting relevant noncoding SNVs in the mouse.
Collapse
Affiliation(s)
- Pan Gao
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Qing Lyu
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Amr R. Ghanam
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Cicera R. Lazzarotto
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38195 USA
| | - Gregory A. Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138 USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138 USA
| | - Wei Zhang
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Mihyun Choi
- Department of Physiology, Albany Medical College, Albany, NY 12208 USA
| | - Orazio J. Slivano
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Kevin Holden
- Synthego Corporation, Redwood City, CA 94025 USA
| | | | | | - Rob J. Munroe
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853 USA
| | | | - John C. Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853 USA
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138 USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138 USA
| | - Shengdar Q. Tsai
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38195 USA
| | - Xiaochun Long
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | - Joseph M. Miano
- Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| |
Collapse
|
6
|
Rippe C, Morén B, Liu L, Stenkula KG, Mustaniemi J, Wennström M, Swärd K. NG2/CSPG4, CD146/MCAM and VAP1/AOC3 are regulated by myocardin-related transcription factors in smooth muscle cells. Sci Rep 2021; 11:5955. [PMID: 33727640 PMCID: PMC7966398 DOI: 10.1038/s41598-021-85335-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/26/2021] [Indexed: 12/28/2022] Open
Abstract
The present work addressed the hypothesis that NG2/CSPG4, CD146/MCAM, and VAP1/AOC3 are target genes of myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MKL1, MRTF-B/MKL2) and serum response factor (SRF). Using a bioinformatics approach, we found that CSPG4, MCAM, and AOC3 correlate with MYOCD, MRTF-A/MKL1, and SRF across human tissues. No other transcription factor correlated as strongly with these transcripts as SRF. Overexpression of MRTFs increased both mRNA and protein levels of CSPG4, MCAM, and AOC3 in cultured human smooth muscle cells (SMCs). Imaging confirmed increased staining for CSPG4, MCAM, and AOC3 in MRTF-A/MKL1-transduced cells. MRTFs exert their effects through SRF, and the MCAM and AOC3 gene loci contained binding sites for SRF. SRF silencing reduced the transcript levels of these genes, and time-courses of induction paralleled the direct target ACTA2. MRTF-A/MKL1 increased the activity of promoter reporters for MCAM and AOC3, and transcriptional activation further depended on the chromatin remodeling enzyme KDM3A. CSPG4, MCAM, and AOC3 responded to the MRTF-SRF inhibitor CCG-1423, to actin dynamics, and to ternary complex factors. Coincidental detection of these proteins should reflect MRTF-SRF activity, and beyond SMCs, we observed co-expression of CD146/MCAM, NG2/CSPG4, and VAP1/AOC3 in pericytes and endothelial cells in the human brain. This work identifies highly responsive vascular target genes of MRTF-SRF signaling that are regulated via a mechanism involving KDM3A.
Collapse
Affiliation(s)
- Catarina Rippe
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Björn Morén
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Li Liu
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.,Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Karin G Stenkula
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Johan Mustaniemi
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Malin Wennström
- Department of Clinical Sciences, Malmö, Lund University, 221 84, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
7
|
Zhu B, Daoud F, Zeng S, Matic L, Hedin U, Uvelius B, Rippe C, Albinsson S, Swärd K. Antagonistic relationship between the unfolded protein response and myocardin-driven transcription in smooth muscle. J Cell Physiol 2020; 235:7370-7382. [PMID: 32039481 DOI: 10.1002/jcp.29637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/30/2020] [Indexed: 01/28/2023]
Abstract
Smooth muscle cells (SMCs) are characterized by a high degree of phenotypic plasticity. Contractile differentiation is governed by myocardin-related transcription factors (MRTFs), in particular myocardin (MYOCD), and when their drive is lost, the cells become proliferative and synthetic with an expanded endoplasmic reticulum (ER). ER is responsible for assembly and folding of secreted proteins. When the load on the ER surpasses its capacity, three stress sensors (activating transcription factor 6 [ATF6], inositol-requiring enzyme 1α [IRE1α]/X-box binding protein 1 [XBP1], and PERK/ATF4) are activated to expand the ER and increase its folding capacity. This is referred to as the unfolded protein response (UPR). Here, we hypothesized that there is a reciprocal relationship between SMC differentiation and the UPR. Tight negative correlations between SMC markers (MYH11, MYOCD, KCNMB1, SYNPO2) and UPR markers (SDF2L1, CALR, MANF, PDIA4) were seen in microarray data sets from carotid arterial injury, partial bladder outlet obstruction, and bladder denervation, respectively. The UPR activators dithiothreitol (DTT) and tunicamycin (TN) activated the UPR and reduced MYOCD along with SMC markers in vitro. The IRE1α inhibitor 4μ8C counteracted the effect of DTT and TN on SMC markers and MYOCD expression. Transfection of active XBP1s was sufficient to reduce both MYOCD and the SMC markers. MRTFs also antagonized the UPR as indicated by reduced TN and DTT-mediated induction of CRELD2, MANF, PDIA4, and SDF2L1 following overexpression of MRTFs. The latter effect did not involve the newly identified MYOCD/SRF target MSRB3, or reduced production of either XBP1s or cleaved ATF6. The UPR thus counteracts SMC differentiation via the IRE1α/XBP1 arm of the UPR and MYOCD repression.
Collapse
Affiliation(s)
- Baoyi Zhu
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Guangdong, China.,Department of Experimental Medical Science, Lund, Sweden
| | - Fatima Daoud
- Department of Experimental Medical Science, Lund, Sweden
| | - Shaohua Zeng
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Guangdong, China
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Uvelius
- Department of Clinical Science, Lund University, Lund, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund, Sweden
| | | | - Karl Swärd
- Department of Experimental Medical Science, Lund, Sweden
| |
Collapse
|
8
|
Swärd K, Krawczyk KK, Morén B, Zhu B, Matic L, Holmberg J, Hedin U, Uvelius B, Stenkula K, Rippe C. Identification of the intermediate filament protein synemin/SYNM as a target of myocardin family coactivators. Am J Physiol Cell Physiol 2019; 317:C1128-C1142. [PMID: 31461342 DOI: 10.1152/ajpcell.00047.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myocardin (MYOCD) is a critical regulator of smooth muscle cell (SMC) differentiation, but its transcriptional targets remain to be exhaustively characterized, especially at the protein level. Here we leveraged human RNA and protein expression data to identify novel potential MYOCD targets. Using correlation analyses we found several targets that we could confirm at the protein level, including SORBS1, SLMAP, SYNM, and MCAM. We focused on SYNM, which encodes the intermediate filament protein synemin. SYNM rivalled smooth muscle myosin (MYH11) for SMC specificity and was controlled at the mRNA and protein levels by all myocardin-related transcription factors (MRTFs: MYOCD, MRTF-A/MKL1, and MRTF-B/MKL2). MRTF activity is regulated by the ratio of filamentous to globular actin, and SYNM was accordingly reduced by interventions that depolymerize actin, such as latrunculin treatment and overexpression of constitutively active cofilin. Many MRTF target genes depend on serum response factor (SRF), but SYNM lacked SRF-binding motifs in its proximal promoter, which was not directly regulated by MYOCD. Furthermore, SYNM resisted SRF silencing, yet the time course of induction closely paralleled that of the SRF-dependent target gene ACTA2. SYNM was repressed by the ternary complex factor (TCF) FLI1 and was increased in mouse embryonic fibroblasts lacking three classical TCFs (ELK1, ELK3, and ELK4). Imaging showed colocalization of SYNM with the intermediate filament proteins desmin and vimentin, and MRTF-A/MKL1 increased SYNM-containing intermediate filaments in SMCs. These studies identify SYNM as a novel SRF-independent target of myocardin that is abundantly expressed in all SMCs.
Collapse
Affiliation(s)
- Karl Swärd
- Department of Experimental Medical Science, Lund, Sweden
| | | | - Björn Morén
- Department of Experimental Medical Science, Lund, Sweden
| | - Baoyi Zhu
- Department of Experimental Medical Science, Lund, Sweden.,Department of Urology, the Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Guangdong, China
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Uvelius
- Department of Clinical Science, Lund, Lund University, Lund, Sweden
| | - Karin Stenkula
- Department of Experimental Medical Science, Lund, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund, Sweden
| |
Collapse
|
9
|
Loss of serum response factor in mature neurons in the dentate gyrus alters the morphology of dendritic spines and hippocampus-dependent behavioral tasks. Brain Struct Funct 2019; 224:2691-2701. [PMID: 31375980 PMCID: PMC6778544 DOI: 10.1007/s00429-019-01925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Serum response factor (SRF) is a major transcription factor that regulates the expression of several plasticity-associated genes in the brain. Although the developmental expression of SRF in excitatory neurons is crucial for establishing proper hippocampal circuitry, no substantial evidence of its role in unstimulated mature neurons has been provided. The present study used time-controlled, conditional SRF knockout mice and found that the lack of SRF in adult neurons led to decreased actin levels and inactivation of the actin-severing protein cofilin 1 through its increase in phosphorylation at Ser3. The augmentation of cofilin 1 phosphorylation correlated with an alteration of dendritic spine morphology in the dentate gyrus, which was reflected by an increase in the number of spines that clustered into the long-spine category. The changes in spine morphology coincided with a lower amplitude and frequency of miniature excitatory postsynaptic currents. Moreover, SRF knockout animals were hyperactive and exhibited impairments in hippocampus-dependent behaviors, such as digging, marble burying, and nesting. Altogether, our data indicate that the adult deletion of neuronal SRF leads to alterations of spine morphology and function and hippocampus-dependent behaviors. Thus, SRF deletion in adult neurons recapitulates some aspects of morphological, electrophysiological, and behavioral changes that are observed in such psychiatric disorders as schizophrenia and autism spectrum disorders.
Collapse
|
10
|
Li S, Kvon EZ, Visel A, Pennacchio LA, Ovcharenko I. Stable enhancers are active in development, and fragile enhancers are associated with evolutionary adaptation. Genome Biol 2019; 20:140. [PMID: 31307522 PMCID: PMC6631995 DOI: 10.1186/s13059-019-1750-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
Background Despite continual progress in the identification and characterization of trait- and disease-associated variants that disrupt transcription factor (TF)-DNA binding, little is known about the distribution of TF binding deactivating mutations (deMs) in enhancer sequences. Here, we focus on elucidating the mechanism underlying the different densities of deMs in human enhancers. Results We identify two classes of enhancers based on the density of nucleotides prone to deMs. Firstly, fragile enhancers with abundant deM nucleotides are associated with the immune system and regular cellular maintenance. Secondly, stable enhancers with only a few deM nucleotides are associated with the development and regulation of TFs and are evolutionarily conserved. These two classes of enhancers feature different regulatory programs: the binding sites of pioneer TFs of FOX family are specifically enriched in stable enhancers, while tissue-specific TFs are enriched in fragile enhancers. Moreover, stable enhancers are more tolerant of deMs due to their dominant employment of homotypic TF binding site (TFBS) clusters, as opposed to the larger-extent usage of heterotypic TFBS clusters in fragile enhancers. Notably, the sequence environment and chromatin context of the cognate motif, other than the motif itself, contribute more to the susceptibility to deMs of TF binding. Conclusions This dichotomy of enhancer activity is conserved across different tissues, has a specific footprint in epigenetic profiles, and argues for a bimodal evolution of gene regulatory programs in vertebrates. Specifically encoded stable enhancers are evolutionarily conserved and associated with development, while differently encoded fragile enhancers are associated with the adaptation of species. Electronic supplementary material The online version of this article (10.1186/s13059-019-1750-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shan Li
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Evgeny Z Kvon
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.,United States Department of Energy Joint Genome Institute, Walnut Creek, CA, 94598, USA.,School of Natural Sciences, University of California, Merced, CA, 95343, USA
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.,United States Department of Energy Joint Genome Institute, Walnut Creek, CA, 94598, USA.,Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Ivan Ovcharenko
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
Shabani S, Mashayekhi F, Shahangian SS, Salehi Z. Genetic polymorphism of glial cell-derived neurotrophic factor (GDNF) in male infertility. Br J Biomed Sci 2018; 76:86-88. [DOI: 10.1080/09674845.2018.1545553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- S Shabani
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - F Mashayekhi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - SS Shahangian
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Z Salehi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
12
|
Gau D, Roy P. SRF'ing and SAP'ing - the role of MRTF proteins in cell migration. J Cell Sci 2018; 131:131/19/jcs218222. [PMID: 30309957 DOI: 10.1242/jcs.218222] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Actin-based cell migration is a fundamental cellular activity that plays a crucial role in a wide range of physiological and pathological processes. An essential feature of the remodeling of actin cytoskeleton during cell motility is the de novo synthesis of factors involved in the regulation of the actin cytoskeleton and cell adhesion in response to growth-factor signaling, and this aspect of cell migration is critically regulated by serum-response factor (SRF)-mediated gene transcription. Myocardin-related transcription factors (MRTFs) are key coactivators of SRF that link actin dynamics to SRF-mediated gene transcription. In this Review, we provide a comprehensive overview of the role of MRTF in both normal and cancer cell migration by discussing its canonical SRF-dependent as well as its recently emerged SRF-independent functions, exerted through its SAP domain, in the context of cell migration. We conclude by highlighting outstanding questions for future research in this field.
Collapse
Affiliation(s)
- David Gau
- Department of Bioengineering, University of Pittsburgh, PA 15213, USA
| | - Partha Roy
- Department of Bioengineering, University of Pittsburgh, PA 15213, USA .,Department of Pathology, University of Pittsburgh, PA, 15213, USA
| |
Collapse
|
13
|
Zhu B, Rippe C, Holmberg J, Zeng S, Perisic L, Albinsson S, Hedin U, Uvelius B, Swärd K. Nexilin/NEXN controls actin polymerization in smooth muscle and is regulated by myocardin family coactivators and YAP. Sci Rep 2018; 8:13025. [PMID: 30158653 PMCID: PMC6115340 DOI: 10.1038/s41598-018-31328-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/17/2018] [Indexed: 01/03/2023] Open
Abstract
Nexilin, encoded by the NEXN gene, is expressed in striated muscle and localizes to Z-discs, influencing mechanical stability. We examined Nexilin/NEXN in smooth muscle cells (SMCs), and addressed if Nexilin localizes to dense bodies and dense bands and whether it is regulated by actin-controlled coactivators from the MRTF (MYOCD, MKL1, MKL2) and YAP/TAZ (YAP1 and WWTR1) families. NEXN expression in SMCs was comparable to that in striated muscles. Immunofluorescence and immunoelectron microscopy suggested that Nexilin localizes to dense bodies and dense bands. Correlations at the mRNA level suggested that NEXN expression might be controlled by actin polymerization. Depolymerization of actin using Latrunculin B repressed the NEXN mRNA and protein in bladder and coronary artery SMCs. Overexpression and knockdown supported involvement of both YAP/TAZ and MRTFs in the transcriptional control of NEXN. YAP/TAZ and MRTFs appeared equally important in bladder SMCs, whereas MRTFs dominated in vascular SMCs. Expression of NEXN was moreover reduced in situations of SMC phenotypic modulation in vivo. The proximal promoter of NEXN conferred control by MRTF-A/MKL1 and MYOCD. NEXN silencing reduced actin polymerization and cell migration, as well as SMC marker expression. NEXN targeting by actin-controlled coactivators thus amplifies SMC differentiation through the actin cytoskeleton, probably via dense bodies and dense bands.
Collapse
Affiliation(s)
- Baoyi Zhu
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden. .,Department of Urology, the Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), 511518, Guangdong, China.
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Shaohua Zeng
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Department of Urology, the Sixth Affiliated Hospital of Guangzhou Medical University (Qingyuan People's Hospital), 511518, Guangdong, China
| | - Ljubica Perisic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Uvelius
- Department of Clinical Science, Section of Urology, Lund University, SE-221 84, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| |
Collapse
|
14
|
Zhang X, Azhar G, Wei JY. SIRT2 gene has a classic SRE element, is a downstream target of serum response factor and is likely activated during serum stimulation. PLoS One 2017; 12:e0190011. [PMID: 29267359 PMCID: PMC5739444 DOI: 10.1371/journal.pone.0190011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 12/06/2017] [Indexed: 01/13/2023] Open
Abstract
The sirtuin proteins are an evolutionarily conserved family of NAD+-dependent deacetylases that regulate various cellular functions. Among the seven sirtuins, SIRT2 is predominantly found in the cytoplasm, and is present in a wide range of tissues. Recent studies indicate that SIRT2 plays an important role in metabolic homeostasis. Several studies indicate that SIRT2 is upregulated under serum deprivation conditions. Since the serum response factor gene usually responds rapidly to serum deprivation and/or serum restoration following deprivation, we hypothesized that a common mechanism may serve to regulate both SIRT2 and SRF during serum stimulation. Using a bioinformatics approach, we searched the SRF binding motif in the SIRT2 gene, and found one classic CArG element (CCATAATAGG) in the SIRT2 gene promoter, which was bound to SRF in the electrophoretic mobility shift assay (EMSA). Serum deprivation induced SIRT2 expression, while SRF and the SRF binding protein, p49/STRAP, repressed SIRT2 gene expression. SIRT2 gene expression was also repressed by the Rho/SRF inhibitor, CCG-1423. These data demonstrate that the classic SRE element in the SIRT2 gene promoter region is functional and therefore, SIRT2 gene is a downstream target of the Rho/SRF signaling pathway. The increased expression of SRF that was observed in the aged heart may affect SIRT2 gene expression and contribute to altered metabolic status in senescence.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Gohar Azhar
- Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jeanne Y. Wei
- Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Guo B, Lyu Q, Slivano OJ, Dirkx R, Christie CK, Czyzyk J, Hezel AF, Gharavi AG, Small EM, Miano JM. Serum Response Factor Is Essential for Maintenance of Podocyte Structure and Function. J Am Soc Nephrol 2017; 29:416-422. [PMID: 29114040 DOI: 10.1681/asn.2017050473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/11/2017] [Indexed: 01/15/2023] Open
Abstract
Podocytes contain an intricate actin cytoskeleton that is essential for the specialized function of this cell type in renal filtration. Serum response factor (SRF) is a master transcription factor for the actin cytoskeleton, but the in vivo expression and function of SRF in podocytes are unknown. We found that SRF protein colocalizes with podocyte markers in human and mouse kidneys. Compared with littermate controls, mice in which the Srf gene was conditionally inactivated with NPHS2-Cre exhibited early postnatal proteinuria, hypoalbuminemia, and azotemia. Histologic changes in the mutant mice included glomerular capillary dilation and mild glomerulosclerosis, with reduced expression of multiple canonical podocyte markers. We also noted tubular dilation, cell proliferation, and protein casts as well as reactive changes in mesangial cells and interstitial inflammation. Ultrastructure analysis disclosed foot process effacement with loss of slit diaphragms. To ascertain the importance of SRF cofactors in podocyte function, we disabled the myocardin-related transcription factor A and B genes. Although loss of either SRF cofactor alone had no observable effect in the kidney, deficiency of both recapitulated the Srf-null phenotype. These results establish a vital role for SRF and two SRF cofactors in the maintenance of podocyte structure and function.
Collapse
Affiliation(s)
- Bing Guo
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York.,Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Lyu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Orazio J Slivano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Ronald Dirkx
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Christine K Christie
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Jan Czyzyk
- Department of Pathology and Laboratory Medicine and
| | - Aram F Hezel
- James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Ali G Gharavi
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York;
| |
Collapse
|
16
|
D'Amico F, Skarmoutsou E, Lo LJ, Granata M, Trovato C, Rossi GA, Bellocchi C, Marchini M, Scorza R, Mazzarino MC, Keinan A. Association between rs2294020 in X-linked CCDC22 and susceptibility to autoimmune diseases with focus on systemic lupus erythematosus. Immunol Lett 2016; 181:58-62. [PMID: 27888057 DOI: 10.1016/j.imlet.2016.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/20/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022]
Abstract
Autoimmune diseases often share common susceptibility genes. Most genetic variants associated with susceptibility to systemic lupus erythematosus are also associated with other autoimmune diseases. The X-linked variant rs2294020 is positioned in exon 7 of the CCDC22 gene. The encoded protein functions in the regulation of NF-κB, a master regulator in immune response. The aim of this study is to investigate whether the rs2294020 polymorphism may be a general susceptibility factor for autoimmunity. We evaluated case-control association between the occurrence of rs2294020 and different autoimmune diseases, including new data for systemic lupus erythematosus and previous genome-wide association studies (GWAS) (though most did not analyse the X chromosome) of psoriasis, celiac disease, Crohn's disease, ulcerative colitis, multiple sclerosis, vitiligo, type-1 diabetes, rheumatoid arthritis, and ankylosing spondylitis. Cases from patients affected by amyotrophic lateral sclerosis and type-2 diabetes were also included in the study. We detected nominal significant associations of rs2294020 with systemic lupus erythematosus (additive model test: p=0.01), vitiligo (p=0.016), psoriasis (p=0.038), and in only one of two studies of multiple sclerosis (p=0.03). Our results suggest that rs2294020 is associated with the risk of several autoimmune diseases in European populations, specifically with diseases that present themselves, among else, in the skin.
Collapse
Affiliation(s)
- Fabio D'Amico
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy.
| | - Evangelia Skarmoutsou
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy
| | - Lauren J Lo
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
| | - Mariagrazia Granata
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy
| | - Chiara Trovato
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy
| | - Giulio A Rossi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy
| | - Chiara Bellocchi
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via Pace 9, I-20122 Milan, Italy
| | - Maurizio Marchini
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via Pace 9, I-20122 Milan, Italy
| | - Raffaella Scorza
- Referral Center for Systemic Autoimmune Diseases, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via Pace 9, I-20122 Milan, Italy
| | - Maria Clorinda Mazzarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Androne 83, I-95124 Catania, Italy
| | - Alon Keinan
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
17
|
Chettimada S, Joshi SR, Dhagia V, Aiezza A, Lincoln TM, Gupte R, Miano JM, Gupte SA. Vascular smooth muscle cell contractile protein expression is increased through protein kinase G-dependent and -independent pathways by glucose-6-phosphate dehydrogenase inhibition and deficiency. Am J Physiol Heart Circ Physiol 2016; 311:H904-H912. [PMID: 27521420 PMCID: PMC5114469 DOI: 10.1152/ajpheart.00335.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/06/2016] [Indexed: 11/22/2022]
Abstract
Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-β-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Blotting, Western
- Cattle
- Chromatography, Liquid
- Contractile Proteins/drug effects
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinase Type I/metabolism
- Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinases/metabolism
- Gene Knockdown Techniques
- Glucosephosphate Dehydrogenase/antagonists & inhibitors
- Glucosephosphate Dehydrogenase/genetics
- Immunoprecipitation
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/drug effects
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- MicroRNAs/drug effects
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymerase Chain Reaction
- Rats
- Serum Response Factor/drug effects
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Tandem Mass Spectrometry
- Trans-Activators/drug effects
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Sukrutha Chettimada
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Sachindra Raj Joshi
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Pharmacology, New York Medical College, Valhalla, New York
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | | | - Rakhee Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Sachin A Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
18
|
Zhao J, Zhang W, Lin M, Wu W, Jiang P, Tou E, Xue M, Richards A, Jourd'heuil D, Asif A, Zheng D, Singer HA, Miano JM, Long X. MYOSLID Is a Novel Serum Response Factor-Dependent Long Noncoding RNA That Amplifies the Vascular Smooth Muscle Differentiation Program. Arterioscler Thromb Vasc Biol 2016; 36:2088-99. [PMID: 27444199 DOI: 10.1161/atvbaha.116.307879] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/05/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNA) represent a growing class of noncoding genes with diverse cellular functions. We previously reported on SENCR, an lncRNA that seems to support the vascular smooth muscle cell (VSMC) contractile phenotype. However, information about the VSMC-specific lncRNAs regulated by myocardin (MYOCD)/serum response factor, the master switch for VSMC differentiation, is unknown. APPROACH AND RESULTS To define novel lncRNAs with functions related to VSMC differentiation, we performed RNA sequencing in human coronary artery SMCs that overexpress MYOCD. Several novel lncRNAs showed altered expression with MYOCD overexpression and one, named MYOcardin-induced Smooth muscle LncRNA, Inducer of Differentiation (MYOSLID), was activated by MYOCD and selectively expressed in VSMCs. MYOSLID was a direct transcriptional target of both MYOCD/serum response factor and transforming growth factor-β/SMAD pathways. Functional studies revealed that MYOSLID promotes VSMC differentiation and inhibits VSMC proliferation. MYOSLID showed reduced expression in failed human arteriovenous fistula samples compared with healthy veins. Although MYOSLID did not affect gene expression of transcription factors, such as serum response factor and MYOCD, its depletion in VSMCs disrupted actin stress fiber formation and blocked nuclear translocation of MYOCD-related transcription factor A (MKL1). Finally, loss of MYOSLID abrogated transforming growth factor-β1-induced SMAD2 phosphorylation. CONCLUSIONS We have demonstrated that MYOSLID, the first human VSMC-selective and serum response factor/CArG-dependent lncRNA, is a novel modulator in amplifying the VSMC differentiation program, likely through feed-forward actions of both MKL1 and transforming growth factor-β/SMAD pathways.
Collapse
Affiliation(s)
- Jinjing Zhao
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Wei Zhang
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Mingyan Lin
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Wen Wu
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Pengtao Jiang
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Emiley Tou
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Min Xue
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Angelene Richards
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - David Jourd'heuil
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Arif Asif
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Deyou Zheng
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Harold A Singer
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Joseph M Miano
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.)
| | - Xiaochun Long
- From the Department of Molecular and Cellular Physiology (J.Z., W.Z., W.W., E.T., M.X., A.R., D.J., H.A.S., X.L.), Albany Medical College, NY; Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ (A.A.); Departments of Genetics (M.L., D.Z.) and Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (P.J., J.M.M.); and National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, P.R. China (M.X.).
| |
Collapse
|
19
|
Miano JM, Zhu QM, Lowenstein CJ. A CRISPR Path to Engineering New Genetic Mouse Models for Cardiovascular Research. Arterioscler Thromb Vasc Biol 2016; 36:1058-75. [PMID: 27102963 PMCID: PMC4882230 DOI: 10.1161/atvbaha.116.304790] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/06/2016] [Indexed: 12/26/2022]
Abstract
Previous efforts to target the mouse genome for the addition, subtraction, or substitution of biologically informative sequences required complex vector design and a series of arduous steps only a handful of laboratories could master. The facile and inexpensive clustered regularly interspaced short palindromic repeats (CRISPR) method has now superseded traditional means of genome modification such that virtually any laboratory can quickly assemble reagents for developing new mouse models for cardiovascular research. Here, we briefly review the history of CRISPR in prokaryotes, highlighting major discoveries leading to its formulation for genome modification in the animal kingdom. Core components of CRISPR technology are reviewed and updated. Practical pointers for 2-component and 3-component CRISPR editing are summarized with many applications in mice including frameshift mutations, deletion of enhancers and noncoding genes, nucleotide substitution of protein-coding and gene regulatory sequences, incorporation of loxP sites for conditional gene inactivation, and epitope tag integration. Genotyping strategies are presented and topics of genetic mosaicism and inadvertent targeting discussed. Finally, clinical applications and ethical considerations are addressed as the biomedical community eagerly embraces this astonishing innovation in genome editing to tackle previously intractable questions.
Collapse
Affiliation(s)
- Joseph M Miano
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (J.M.M., Q.M.Z., C.J.L.); and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA (Q.M.Z.).
| | - Qiuyu Martin Zhu
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (J.M.M., Q.M.Z., C.J.L.); and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA (Q.M.Z.)
| | - Charles J Lowenstein
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (J.M.M., Q.M.Z., C.J.L.); and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA (Q.M.Z.)
| |
Collapse
|
20
|
Ro S. Multi-phenotypic Role of Serum Response Factor in the Gastrointestinal System. J Neurogastroenterol Motil 2016; 22:193-200. [PMID: 26727951 PMCID: PMC4819857 DOI: 10.5056/jnm15183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/26/2015] [Indexed: 12/12/2022] Open
Abstract
Serum response factor (SRF) is a master transcription factor of the actin cytoskeleton that binds to highly conserved CArG boxes located within the majority of smooth muscle cell (SMC)-restricted promoters/enhancers. Although most studies of SRF focus on skeletal muscle, cardiac muscle, and vascular SMCs, SRF research has recently expanded into the gastrointestinal (GI) system. Genome scale analyses of GI SMC transcriptome and CArG boxes (CArGome) have identified new SRF target genes. In addition to circular and longitudinal smooth muscle layers, SRF is also expressed in GI mucosa and cancers. In the GI tract, SRF is the central regulator of genes involved in apoptosis, dedifferentiation, proliferation, and migration of cells. Since SRF is the cell phenotypic modulator, it may play an essential role in the development of myopathy, hypertrophy, ulcers, gastric and colon cancers within the GI tract. Given the multi-functional role displayed by SRF in the digestive system, SRF has received more attention emerging as a potential therapeutic target. This review summarizes the findings in SRF research pertaining to the GI tract and provides valuable insight into future directions.
Collapse
Affiliation(s)
- Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
21
|
Kuzniewska B, Nader K, Dabrowski M, Kaczmarek L, Kalita K. Adult Deletion of SRF Increases Epileptogenesis and Decreases Activity-Induced Gene Expression. Mol Neurobiol 2016; 53:1478-1493. [PMID: 25636686 PMCID: PMC4789231 DOI: 10.1007/s12035-014-9089-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/29/2014] [Indexed: 11/27/2022]
Abstract
Although the transcription factor serum response factor (SRF) has been suggested to play a role in activity-dependent gene expression and mediate plasticity-associated structural changes in the hippocampus, no unequivocal evidence has been provided for its role in brain pathology, such as epilepsy. A genome-wide program of activity-induced genes that are regulated by SRF also remains unknown. In the present study, we show that the inducible and conditional deletion of SRF in the adult mouse hippocampus increases the epileptic phenotype in the kainic acid model of epilepsy, reflected by more severe and frequent seizures. Moreover, we observe a robust decrease in activity-induced gene transcription in SRF knockout mice. We characterize the genetic program controlled by SRF in neurons and using functional annotation, we find that SRF target genes are associated with synaptic plasticity and epilepsy. Several of these SRF targets function as regulators of inhibitory or excitatory balance and the structural plasticity of neurons. Interestingly, mutations in those SRF targets have found to be associated with such human neuropsychiatric disorders, as autism and intellectual disability. We also identify novel direct SRF targets in hippocampus: Npas4, Gadd45g, and Zfp36. Altogether, our data indicate that proteins that are highly upregulated by neuronal stimulation, identified in the present study as SRF targets, may function as endogenous protectors against overactivation. Thus, the lack of these effector proteins in SRF knockout animals may lead to uncontrolled excitation and eventually epilepsy.
Collapse
Affiliation(s)
- Bozena Kuzniewska
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Karolina Nader
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Neurobiology Center, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland.
| |
Collapse
|
22
|
Lighthouse JK, Small EM. Transcriptional control of cardiac fibroblast plasticity. J Mol Cell Cardiol 2016; 91:52-60. [PMID: 26721596 PMCID: PMC4764462 DOI: 10.1016/j.yjmcc.2015.12.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/15/2015] [Accepted: 12/20/2015] [Indexed: 12/11/2022]
Abstract
Cardiac fibroblasts help maintain the normal architecture of the healthy heart and are responsible for scar formation and the healing response to pathological insults. Various genetic, biomechanical, or humoral factors stimulate fibroblasts to become contractile smooth muscle-like cells called myofibroblasts that secrete large amounts of extracellular matrix. Unfortunately, unchecked myofibroblast activation in heart disease leads to pathological fibrosis, which is a major risk factor for the development of cardiac arrhythmias and heart failure. A better understanding of the molecular mechanisms that control fibroblast plasticity and myofibroblast activation is essential to develop novel strategies to specifically target pathological cardiac fibrosis without disrupting the adaptive healing response. This review highlights the major transcriptional mediators of fibroblast origin and function in development and disease. The contribution of the fetal epicardial gene program will be discussed in the context of fibroblast origin in development and following injury, primarily focusing on Tcf21 and C/EBP. We will also highlight the major transcriptional regulatory axes that control fibroblast plasticity in the adult heart, including transforming growth factor β (TGFβ)/Smad signaling, the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) axis, and Calcineurin/transient receptor potential channel (TRP)/nuclear factor of activated T-Cell (NFAT) signaling. Finally, we will discuss recent strategies to divert the fibroblast transcriptional program in an effort to promote cardiomyocyte regeneration. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling".
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA.
| |
Collapse
|
23
|
Human Genes Encoding Transcription Factors and Chromatin-Modifying Proteins Have Low Levels of Promoter Polymorphism: A Study of 1000 Genomes Project Data. Int J Genomics 2015; 2015:260159. [PMID: 26417590 PMCID: PMC4568383 DOI: 10.1155/2015/260159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/29/2015] [Indexed: 12/15/2022] Open
Abstract
The expression level of each gene is controlled by its regulatory regions, which determine the precise regulation in a tissue-specific manner, according to the developmental stage of the body and the necessity of a response to external stimuli. Nucleotide substitutions in regulatory gene regions may modify the affinity of transcription factors to their specific DNA binding sites, affecting the transcription rates of genes. In our previous research, we found that genes controlling the sensory perception of smell and genes involved in antigen processing and presentation were overrepresented significantly among genes with high SNP contents in their promoter regions. The goal of our study was to reveal functional features of human genes containing extremely small numbers of SNPs in promoter regions. Two functional groups were found to be overrepresented among genes whose promoters did not contain SNPs: (1) genes involved in gene-specific transcription and (2) genes controlling chromatin organization. We revealed that the 5′-regulatory regions of genes encoding transcription factors and chromatin-modifying proteins were characterized by reduced genetic variability. One important exception from this rule refers to genes encoding transcription factors with zinc-coordinating DNA-binding domains (DBDs), which underwent extensive expansion in vertebrates, particularly, in primate evolution. Hence, we obtained new evidence for evolutionary forces shaping variability in 5′-regulatory regions of genes.
Collapse
|
24
|
Abstract
Caveolae are membrane organelles that play roles in glucose and lipid metabolism and in vascular function. Formation of caveolae requires caveolins and cavins. The make-up of caveolae and their density is considered to reflect cell-specific transcriptional control mechanisms for caveolins and cavins, but knowledge regarding regulation of caveolae genes is incomplete. Myocardin (MYOCD) and its relative MRTF-A (MKL1) are transcriptional coactivators that control genes which promote smooth muscle differentiation. MRTF-A communicates changes in actin polymerization to nuclear gene transcription. Here we tested if myocardin family proteins control biogenesis of caveolae via activation of caveolin and cavin transcription. Using human coronary artery smooth muscle cells we found that jasplakinolide and latrunculin B (LatB), substances that promote and inhibit actin polymerization, increased and decreased protein levels of caveolins and cavins, respectively. The effect of LatB was associated with reduced mRNA levels for these genes and this was replicated by the MRTF inhibitor CCG-1423 which was non-additive with LatB. Overexpression of myocardin and MRTF-A caused 5-10-fold induction of caveolins whereas cavin-1 and cavin-2 were induced 2-3-fold. PACSIN2 also increased, establishing positive regulation of caveolae genes from three families. Full regulation of CAV1 was retained in its proximal promoter. Knock down of the serum response factor (SRF), which mediates many of the effects of myocardin, decreased cavin-1 but increased caveolin-1 and -2 mRNAs. Viral transduction of myocardin increased the density of caveolae 5-fold in vitro. A decrease of CAV1 was observed concomitant with a decrease of the smooth muscle marker calponin in aortic aneurysms from mice (C57Bl/6) infused with angiotensin II. Human expression data disclosed correlations of MYOCD with CAV1 in a majority of human tissues and in the heart, correlation with MKL2 (MRTF-B) was observed. The myocardin family of transcriptional coactivators therefore drives formation of caveolae and this effect is largely independent of SRF.
Collapse
|
25
|
Lee MY, Park C, Berent RM, Park PJ, Fuchs R, Syn H, Chin A, Townsend J, Benson CC, Redelman D, Shen TW, Park JK, Miano JM, Sanders KM, Ro S. Smooth Muscle Cell Genome Browser: Enabling the Identification of Novel Serum Response Factor Target Genes. PLoS One 2015; 10:e0133751. [PMID: 26241044 PMCID: PMC4524680 DOI: 10.1371/journal.pone.0133751] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/30/2015] [Indexed: 11/18/2022] Open
Abstract
Genome-scale expression data on the absolute numbers of gene isoforms offers essential clues in cellular functions and biological processes. Smooth muscle cells (SMCs) perform a unique contractile function through expression of specific genes controlled by serum response factor (SRF), a transcription factor that binds to DNA sites known as the CArG boxes. To identify SRF-regulated genes specifically expressed in SMCs, we isolated SMC populations from mouse small intestine and colon, obtained their transcriptomes, and constructed an interactive SMC genome and CArGome browser. To our knowledge, this is the first online resource that provides a comprehensive library of all genetic transcripts expressed in primary SMCs. The browser also serves as the first genome-wide map of SRF binding sites. The browser analysis revealed novel SMC-specific transcriptional variants and SRF target genes, which provided new and unique insights into the cellular and biological functions of the cells in gastrointestinal (GI) physiology. The SRF target genes in SMCs, which were discovered in silico, were confirmed by proteomic analysis of SMC-specific Srf knockout mice. Our genome browser offers a new perspective into the alternative expression of genes in the context of SRF binding sites in SMCs and provides a valuable reference for future functional studies.
Collapse
Affiliation(s)
- Moon Young Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, Jeollabuk-do, Korea
| | - Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Robyn M. Berent
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Paul J. Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Robert Fuchs
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Hannah Syn
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Albert Chin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Jared Townsend
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Craig C. Benson
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Doug Redelman
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Tsai-wei Shen
- LC Sciences, 2575 West Bellfort Street Suite 270, Houston, Texas, United States of America
| | - Jong Kun Park
- Division of Biological Science, Wonkwang University, Iksan, Jeollabuk-do, South Korea
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
26
|
Miano JM, Long X. The short and long of noncoding sequences in the control of vascular cell phenotypes. Cell Mol Life Sci 2015; 72:3457-88. [PMID: 26022065 DOI: 10.1007/s00018-015-1936-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022]
Abstract
The two principal cell types of importance for normal vessel wall physiology are smooth muscle cells and endothelial cells. Much progress has been made over the past 20 years in the discovery and function of transcription factors that coordinate proper differentiation of these cells and the maintenance of vascular homeostasis. More recently, the converging fields of bioinformatics, genomics, and next generation sequencing have accelerated discoveries in a number of classes of noncoding sequences, including transcription factor binding sites (TFBS), microRNA genes, and long noncoding RNA genes, each of which mediates vascular cell differentiation through a variety of mechanisms. Alterations in the nucleotide sequence of key TFBS or deviations in transcription of noncoding RNA genes likely have adverse effects on normal vascular cell phenotype and function. Here, the subject of noncoding sequences that influence smooth muscle cell or endothelial cell phenotype will be summarized as will future directions to further advance our understanding of the increasingly complex molecular circuitry governing normal vascular cell differentiation and how such information might be harnessed to combat vascular diseases.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA,
| | | |
Collapse
|
27
|
Vengrenyuk Y, Nishi H, Long X, Ouimet M, Savji N, Martinez FO, Cassella CP, Moore KJ, Ramsey SA, Miano JM, Fisher EA. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler Thromb Vasc Biol 2015; 35:535-46. [PMID: 25573853 PMCID: PMC4344402 DOI: 10.1161/atvbaha.114.304029] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We previously showed that cholesterol loading in vitro converts mouse aortic vascular smooth muscle cells (VSMC) from a contractile state to one resembling macrophages. In human and mouse atherosclerotic plaques, it has become appreciated that ≈40% of cells classified as macrophages by histological markers may be of VSMC origin. Therefore, we sought to gain insight into the molecular regulation of this clinically relevant process. APPROACH AND RESULTS VSMC of mouse (or human) origin were incubated with cyclodextrin-cholesterol complexes for 72 hours, at which time the expression at the protein and mRNA levels of contractile-related proteins was reduced and of macrophage markers increased. Concurrent was downregulation of miR-143/145, which positively regulate the master VSMC differentiation transcription factor myocardin. Mechanisms were further probed in mouse VSMC. Maintaining the expression of myocardin or miR-143/145 prevented and reversed phenotypic changes caused by cholesterol loading. Reversal was also seen when cholesterol efflux was stimulated after loading. Notably, despite expression of macrophage markers, bioinformatic analyses showed that cholesterol-loaded cells remained closer to the VSMC state, consistent with impairment in classical macrophage functions of phagocytosis and efferocytosis. In apoE-deficient atherosclerotic plaques, cells positive for VSMC and macrophage markers were found lining the cholesterol-rich necrotic core. CONCLUSIONS Cholesterol loading of VSMC converts them to a macrophage-appearing state by downregulating the miR-143/145-myocardin axis. Although these cells would be classified by immunohistochemistry as macrophages in human and mouse plaques, their transcriptome and functional properties imply that their contributions to atherogenesis would not be those of classical macrophages.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Binding Sites
- Cell Lineage
- Cell Transdifferentiation
- Cholesterol/metabolism
- Cholesterol, HDL/metabolism
- Coculture Techniques
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Humans
- Jurkat Cells
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Phagocytosis
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Sterol Regulatory Element Binding Protein 2/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
Collapse
Affiliation(s)
- Yuliya Vengrenyuk
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Hitoo Nishi
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Xiaochun Long
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Mireille Ouimet
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nazir Savji
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Fernando O Martinez
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Courtney P Cassella
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Stephen A Ramsey
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|
28
|
Han Y, Slivano OJ, Christie CK, Cheng AW, Miano JM. CRISPR-Cas9 genome editing of a single regulatory element nearly abolishes target gene expression in mice--brief report. Arterioscler Thromb Vasc Biol 2015; 35:312-5. [PMID: 25538209 PMCID: PMC4304932 DOI: 10.1161/atvbaha.114.305017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To ascertain the importance of a single regulatory element in the control of Cnn1 expression using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genome editing. APPROACH AND RESULTS The CRISPR/Cas9 system was used to produce 3 of 18 founder mice carrying point mutations in an intronic CArG box of the smooth muscle cell-restricted Cnn1 gene. Each founder was bred for germline transmission of the mutant CArG box and littermate interbreeding to generate homozygous mutant (Cnn1(ΔCArG/ΔCArG)) mice. Quantitative reverse transcription polymerase chain reaction, Western blotting, and confocal immunofluorescence microscopy showed dramatic reductions in Cnn1 mRNA and CNN1 protein expression in Cnn1(ΔCArG/ΔCArG) mice with no change in other smooth muscle cell-restricted genes and little evidence of off-target edits elsewhere in the genome. In vivo chromatin immunoprecipitation assay revealed a sharp decrease in binding of serum response factor to the mutant CArG box. Loss of CNN1 expression was coincident with an increase in Ki-67 positive cells in the normal vessel wall. CONCLUSIONS CRISPR/Cas9 genome editing of a single CArG box nearly abolishes Cnn1 expression in vivo and evokes increases in smooth muscle cell DNA synthesis. This facile genome editing system paves the way for a new generation of studies designed to test the importance of individual regulatory elements in living animals, including regulatory variants in conserved sequence blocks linked to human disease.
Collapse
Affiliation(s)
- Yu Han
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Orazio J Slivano
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Christine K Christie
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Albert W Cheng
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Joseph M Miano
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.).
| |
Collapse
|
29
|
Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem 2014; 289:33629-43. [PMID: 25301952 DOI: 10.1074/jbc.m114.589556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.
Collapse
Affiliation(s)
- Sarah E Brennan-Laun
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and
| | - Xiao-Ling Li
- the Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather J Ezelle
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| | | | - Perry J Blackshear
- the Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Gerald M Wilson
- From the Marlene and Stewart Greenebaum Cancer Center, Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Bret A Hassel
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| |
Collapse
|
30
|
Ignatieva EV, Levitsky VG, Yudin NS, Moshkin MP, Kolchanov NA. Genetic basis of olfactory cognition: extremely high level of DNA sequence polymorphism in promoter regions of the human olfactory receptor genes revealed using the 1000 Genomes Project dataset. Front Psychol 2014; 5:247. [PMID: 24715883 PMCID: PMC3970011 DOI: 10.3389/fpsyg.2014.00247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 03/05/2014] [Indexed: 11/13/2022] Open
Abstract
The molecular mechanism of olfactory cognition is very complicated. Olfactory cognition is initiated by olfactory receptor proteins (odorant receptors), which are activated by olfactory stimuli (ligands). Olfactory receptors are the initial player in the signal transduction cascade producing a nerve impulse, which is transmitted to the brain. The sensitivity to a particular ligand depends on the expression level of multiple proteins involved in the process of olfactory cognition: olfactory receptor proteins, proteins that participate in signal transduction cascade, etc. The expression level of each gene is controlled by its regulatory regions, and especially, by the promoter [a region of DNA about 100–1000 base pairs long located upstream of the transcription start site (TSS)]. We analyzed single nucleotide polymorphisms using human whole-genome data from the 1000 Genomes Project and revealed an extremely high level of single nucleotide polymorphisms in promoter regions of olfactory receptor genes and HLA genes. We hypothesized that the high level of polymorphisms in olfactory receptor promoters was responsible for the diversity in regulatory mechanisms controlling the expression levels of olfactory receptor proteins. Such diversity of regulatory mechanisms may cause the great variability of olfactory cognition of numerous environmental olfactory stimuli perceived by human beings (air pollutants, human body odors, odors in culinary etc.). In turn, this variability may provide a wide range of emotional and behavioral reactions related to the vast variety of olfactory stimuli.
Collapse
Affiliation(s)
- Elena V Ignatieva
- Laboratory of Evolutionary Bioinformatics and Theoretical Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia ; Department of Natural Science, Novosibirsk State University Novosibirsk, Russia
| | - Victor G Levitsky
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Molecular-Genetic Systems, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Nikolay S Yudin
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Human Molecular Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Mikhail P Moshkin
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Laboratory of Mammalian Ecological Genetics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Department of Natural Science, Novosibirsk State University Novosibirsk, Russia ; Department of Systems Biology, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences Novosibirsk, Russia ; National Research centre "Kurchatov Institute" Moscow, Russia
| |
Collapse
|
31
|
Bell RD, Long X, Lin M, Bergmann JH, Nanda V, Cowan SL, Zhou Q, Han Y, Spector DL, Zheng D, Miano JM. Identification and initial functional characterization of a human vascular cell-enriched long noncoding RNA. Arterioscler Thromb Vasc Biol 2014; 34:1249-59. [PMID: 24578380 DOI: 10.1161/atvbaha.114.303240] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) represent a rapidly growing class of RNA genes with functions related primarily to transcriptional and post-transcriptional control of gene expression. There is a paucity of information about lncRNA expression and function in human vascular cells. Thus, we set out to identify novel lncRNA genes in human vascular smooth muscle cells and to gain insight into their role in the control of smooth muscle cell phenotypes. APPROACH AND RESULTS RNA sequencing (RNA-seq) of human coronary artery smooth muscle cells revealed 31 unannotated lncRNAs, including a vascular cell-enriched lncRNA (Smooth muscle and Endothelial cell-enriched migration/differentiation-associated long NonCoding RNA [SENCR]). Strand-specific reverse transcription polymerase chain reaction (PCR) and rapid amplification of cDNA ends indicate that SENCR is transcribed antisense from the 5' end of the FLI1 gene and exists as 2 splice variants. RNA fluorescence in situ hybridization and biochemical fractionation studies demonstrate SENCR is a cytoplasmic lncRNA. Consistent with this observation, knockdown studies reveal little to no cis-acting effect of SENCR on FLI1 or neighboring gene expression. RNA-seq experiments in smooth muscle cells after SENCR knockdown disclose decreased expression of Myocardin and numerous smooth muscle contractile genes, whereas several promigratory genes are increased. Reverse transcription PCR and Western blotting experiments validate several differentially expressed genes after SENCR knockdown. Loss-of-function studies in scratch wound and Boyden chamber assays support SENCR as an inhibitor of smooth muscle cell migration. CONCLUSIONS SENCR is a new vascular cell-enriched, cytoplasmic lncRNA that seems to stabilize the smooth muscle cell contractile phenotype.
Collapse
Affiliation(s)
- Robert D Bell
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Xiaochun Long
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Mingyan Lin
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Jan H Bergmann
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Vivek Nanda
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Sarah L Cowan
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Qian Zhou
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Yu Han
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - David L Spector
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Deyou Zheng
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.)
| | - Joseph M Miano
- From the Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY (R.D.B., X.L., V.N., S.L.C., Q.Z., Y.H., J.M.M.); Department of Genetics (M.L., D.Z.) and Departments of Neurology and Neuroscience (D.Z.), Albert Einstein College of Medicine, Bronx, NY; and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (J.H.B., D.L.S.).
| |
Collapse
|
32
|
Kawakami-Schulz SV, Verdoni AM, Sattler SG, Jessen E, Kao WWY, Ikeda A, Ikeda S. Serum response factor: positive and negative regulation of an epithelial gene expression network in the destrin mutant cornea. Physiol Genomics 2014; 46:277-89. [PMID: 24550211 DOI: 10.1152/physiolgenomics.00126.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Increased angiogenesis, inflammation, and proliferation are hallmarks of diseased tissues, and in vivo models of these disease phenotypes can provide insight into disease pathology. Dstn(corn1) mice, deficient for the actin depolymerizing factor destrin (DSTN), display an increase of serum response factor (SRF) that results in epithelial hyperproliferation, inflammation, and neovascularization in the cornea. Previous work demonstrated that conditional ablation of Srf from the corneal epithelium of Dstn(corn1) mice returns the cornea to a wild-type (WT) like state. This result implicated SRF as a major regulator of genes that contributes to abnormal phenotypes in Dstn(corn1) cornea. The purpose of this study is to identify gene networks that are affected by increased expression of Srf in the Dstn(corn1) cornea. Microarray analysis led to characterization of gene expression changes that occur when conditional knockout of Srf rescues mutant phenotypes in the cornea of Dstn(corn1) mice. Comparison of gene expression values from WT, Dstn(corn1) mutant, and Dstn(corn1) rescued cornea identified >400 differentially expressed genes that are downstream from SRF. Srf ablation had a significant effect on genes associated with epithelial cell-cell junctions and regulation of actin dynamics. The majority of genes affected by SRF are downregulated in the Dstn(corn1) mutant cornea, suggesting that increased SRF negatively affects transcription of SRF gene targets. ChIP-seq analysis on Dstn(corn1) mutant and WT tissue revealed that, despite being present in higher abundance, SRF binding is significantly decreased in the Dstn(corn1) mutant cornea. This study uses a unique model combining genetic and genomic approaches to identify genes that are regulated by SRF. These findings expand current understanding of the role of SRF in both normal and abnormal tissue homeostasis.
Collapse
|
33
|
Kawakami-Schulz SV, Sattler SG, Doebley AL, Ikeda A, Ikeda S. Genetic modification of corneal neovascularization in Dstn (corn1) mice. Mamm Genome 2013; 24:349-57. [PMID: 23929036 PMCID: PMC3802551 DOI: 10.1007/s00335-013-9468-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/05/2013] [Indexed: 10/26/2022]
Abstract
Mutations in the gene for destrin (Dstn), an actin depolymerizing factor, lead to corneal abnormalities in mice. A null mutation in Dstn, termed Dstn (corn1) , isolated and maintained in the A.BY background (A.BY Dstn (corn1) ), results in corneal epithelial hyperproliferation, inflammation, and neovascularization. We previously reported that neovascularization in the cornea of Dstn (corn1) mice on the C57BL/6 background (B6.A.BY-Dstn (corn1) ) is significantly reduced when compared to A.BY Dstn (corn1) mice, suggesting the existence of genetic modifier(s). The purpose of this study is to identify the genetic basis of the difference in corneal neovascularization between A.BY Dstn (corn1) and B6.A.BY-Dstn (corn1) mice. We generated N2 mice for a whole-genome scan by backcrossing F1 progeny (A.BY Dstn (corn1) × B6.A.BY-Dstn (corn1) ) to B6.A.BY-Dstn (corn1) mice. N2 progeny were quantitatively phenotyped for the extent of corneal neovascularization and genotyped for markers across the mouse genome. We identified significant association of variability in corneal neovascularization with a locus on chromosome 3 (Chr3). The validity of the identified quantitative trait locus (QTL) was tested using B6 consomic mice carrying Chr3 from A/J mice. Dstn (corn1) mice from F1 and F2 intercrosses (B6.A.BY-Dstn (corn1) × C57BL/6J-Chr3(A/J)/NaJ) were phenotyped for the extent of corneal neovascularization. This analysis showed that mice carrying the A/J allele at the QTL show significantly increased neovascularization. Our results indicate the existence of a modifier that genetically interacts with the Dstn gene. This modifier demonstrates allelic differences between C57BL6 and A.BY or A/J. The modifier is sufficient to increase neovascularization in Dstn (corn1) mice.
Collapse
Affiliation(s)
| | - Shannon G. Sattler
- Department of Medical Genetics, University of Wisconsin, Madison, Wisconsin 53706, USA
| | - Anna-Lisa Doebley
- Department of Medical Genetics, University of Wisconsin, Madison, Wisconsin 53706, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin, Madison, Wisconsin 53706, USA
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin, Madison, Wisconsin 53706, USA
| |
Collapse
|
34
|
Martini S, Nair V, Patel SR, Eichinger F, Nelson RG, Weil EJ, Pezzolesi MG, Krolewski AS, Randolph A, Keller BJ, Werner T, Kretzler M. From single nucleotide polymorphism to transcriptional mechanism: a model for FRMD3 in diabetic nephropathy. Diabetes 2013; 62:2605-12. [PMID: 23434934 PMCID: PMC3712052 DOI: 10.2337/db12-1416] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Genome-wide association studies have proven to be highly effective at defining relationships between single nucleotide polymorphisms (SNPs) and clinical phenotypes in complex diseases. Establishing a mechanistic link between a noncoding SNP and the clinical outcome is a significant hurdle in translating associations into biological insight. We demonstrate an approach to assess the functional context of a diabetic nephropathy (DN)-associated SNP located in the promoter region of the gene FRMD3. The approach integrates pathway analyses with transcriptional regulatory pattern-based promoter modeling and allows the identification of a transcriptional framework affected by the DN-associated SNP in the FRMD3 promoter. This framework provides a testable hypothesis for mechanisms of genomic variation and transcriptional regulation in the context of DN. Our model proposes a possible transcriptional link through which the polymorphism in the FRMD3 promoter could influence transcriptional regulation within the bone morphogenetic protein (BMP)-signaling pathway. These findings provide the rationale to interrogate the biological link between FRMD3 and the BMP pathway and serve as an example of functional genomics-based hypothesis generation.
Collapse
Affiliation(s)
- Sebastian Martini
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Viji Nair
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Sanjeevkumar R. Patel
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Felix Eichinger
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Robert G. Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - E. Jennifer Weil
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Marcus G. Pezzolesi
- Research and Clinic Divisions, Joslin Diabetes Center, Boston, Massachusetts
| | | | - Ann Randolph
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin J. Keller
- Department of Computer Science, Eastern Michigan University, Ypsilanti, Michigan
| | - Thomas Werner
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
- Genomatix Software GmbH, Munich, Germany
| | - Matthias Kretzler
- Departments of Internal Medicine and Nephrology, University of Michigan, Ann Arbor, Michigan
- Corresponding author: Matthias Kretzler,
| |
Collapse
|
35
|
Lin C, Hindes A, Burns CJ, Koppel AC, Kiss A, Yin Y, Ma L, Blumenberg M, Khnykin D, Jahnsen FL, Crosby SD, Ramanan N, Efimova T. Serum response factor controls transcriptional network regulating epidermal function and hair follicle morphogenesis. J Invest Dermatol 2012; 133:608-617. [PMID: 23151848 DOI: 10.1038/jid.2012.378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Serum response factor (SRF) is a transcription factor that regulates the expression of growth-related immediate-early, cytoskeletal, and muscle-specific genes to control growth, differentiation, and cytoskeletal integrity in different cell types. To investigate the role for SRF in epidermal development and homeostasis, we conditionally knocked out SRF in epidermal keratinocytes. We report that SRF deletion disrupted epidermal barrier function leading to early postnatal lethality. Mice lacking SRF in epidermis displayed morphogenetic defects, including an eye-open-at-birth phenotype and lack of whiskers. SRF-null skin exhibited abnormal morphology, hyperplasia, aberrant expression of differentiation markers and transcriptional regulators, anomalous actin organization, enhanced inflammation, and retarded hair follicle (HF) development. Transcriptional profiling experiments uncovered profound molecular changes in SRF-null E17.5 epidermis and revealed that many previously identified SRF target CArG box-containing genes were markedly upregulated in SRF-null epidermis, indicating that SRF may function to repress transcription of a subset of its target genes in epidermis. Remarkably, when transplanted onto nude mice, engrafted SRF-null skin lacked hair but displayed normal epidermal architecture with proper expression of differentiation markers, suggesting that although keratinocyte SRF is essential for HF development, a cross-talk between SRF-null keratinocytes and the surrounding microenvironment is likely responsible for the barrier-deficient mutant epidermal phenotype.
Collapse
Affiliation(s)
- Congxing Lin
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Anna Hindes
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Carole J Burns
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Aaron C Koppel
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alexi Kiss
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Yan Yin
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Liang Ma
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Miroslav Blumenberg
- R. O. Perelman Department of Dermatology, NYU School of Medicine, New York, New York, USA
| | - Denis Khnykin
- Department of Pathology and Centre for Immune Regulation, University Hospital and University of Oslo, Oslo, Norway
| | - Frode L Jahnsen
- Department of Pathology and Centre for Immune Regulation, University Hospital and University of Oslo, Oslo, Norway
| | - Seth D Crosby
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Narendrakumar Ramanan
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tatiana Efimova
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
36
|
Expression and promoter analysis of a highly restricted integrin alpha gene in vascular smooth muscle. Gene 2012; 513:82-9. [PMID: 23142384 DOI: 10.1016/j.gene.2012.10.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 09/18/2012] [Accepted: 10/29/2012] [Indexed: 11/19/2022]
Abstract
Full genome annotation requires gene expression analysis and elucidation of promoter activity. Here, we analyzed the expression and promoter of a highly restricted integrin gene, Itga8. RNase protection and quantitative RT-PCR showed Itga8 to be expressed most abundantly in vascular smooth muscle cells (SMC). Transcription start site mapping of Itga8 revealed the immediate 5' promoter region to be poorly conserved with orthologous sequences in the human genome. Further comparative sequence analysis showed a number of conserved non-coding sequence modules around the Itga8 gene. The immediate promoter region and an upstream conserved sequence module were each found to contain a CArG box, which is a binding site for serum response factor (SRF). Luciferase reporter assays revealed activity of several Itga8 promoter constructs with no apparent restricted activity to SMC types. Further, neither SRF nor its coactivator, Myocardin (MYOCD), was able to induce several distinct Itga8 promoter constructs. Transgenic mouse studies failed to reveal Itga8 promoter activity, indicating distal regulatory elements likely control this gene's in vivo expression profile. Interestingly, although the promoter was unresponsive to SRF/MYOCD, the endogenous Itga8 gene showed increases in expression upon ectopic MYOCD expression even though knockdown of SRF both in vitro and in vivo failed to demonstrate a corresponding change in Itga8. Collectively, these data demonstrate that Itga8 expression is CArG-SRF independent, but MYOCD dependent through an as yet unknown sequence module that is distal from the promoter region.
Collapse
|
37
|
Davis J, Burr AR, Davis GF, Birnbaumer L, Molkentin JD. A TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo. Dev Cell 2012; 23:705-15. [PMID: 23022034 DOI: 10.1016/j.devcel.2012.08.017] [Citation(s) in RCA: 272] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 06/05/2012] [Accepted: 08/30/2012] [Indexed: 02/06/2023]
Abstract
After injury or cytokine stimulation, fibroblasts transdifferentiate into myofibroblasts, contractile cells that secrete extracellular matrix for wound healing and tissue remodeling. Here, a genome-wide screen identified TRPC6, a Ca(2+) channel necessary and sufficient for myofibroblast transformation. TRPC6 overexpression fully activated myofibroblast transformation, while fibroblasts lacking Trpc6 were refractory to transforming growth factor β (TGF-β) and angiotensin II-induced transdifferentiation. Trpc6 gene-deleted mice showed impaired dermal and cardiac wound healing after injury. The profibrotic ligands TGF-β and angiotensin II induced TRPC6 expression through p38 mitogen-activated protein kinase (MAPK) serum response factor (SRF) signaling via the TRPC6 promoter. Once induced, TRPC6 activates the Ca(2+)-responsive protein phosphatase calcineurin, which itself induced myofibroblast transdifferentiation. Moreover, inhibition of calcineurin prevented TRPC6-dependent transdifferentiation and dermal wound healing. These results demonstrate an obligate function for TRPC6 and calcineurin in promoting myofibroblast differentiation, suggesting a comprehensive pathway for myofibroblast formation in conjunction with TGF-β, p38 MAPK, and SRF.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
38
|
Serum Response Factor (SRF)-cofilin-actin signaling axis modulates mitochondrial dynamics. Proc Natl Acad Sci U S A 2012; 109:E2523-32. [PMID: 22927399 DOI: 10.1073/pnas.1208141109] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aberrant mitochondrial function, morphology, and transport are main features of neurodegenerative diseases. To date, mitochondrial transport within neurons is thought to rely mainly on microtubules, whereas actin might mediate short-range movements and mitochondrial anchoring. Here, we analyzed the impact of actin on neuronal mitochondrial size and localization. F-actin enhanced mitochondrial size and mitochondrial number in neurites and growth cones. In contrast, raising G-actin resulted in mitochondrial fragmentation and decreased mitochondrial abundance. Cellular F-actin/G-actin levels also regulate serum response factor (SRF)-mediated gene regulation, suggesting a possible link between SRF and mitochondrial dynamics. Indeed, SRF-deficient neurons display neurodegenerative hallmarks of mitochondria, including disrupted morphology, fragmentation, and impaired mitochondrial motility, as well as ATP energy metabolism. Conversely, constitutively active SRF-VP16 induced formation of mitochondrial networks and rescued huntingtin (HTT)-impaired mitochondrial dynamics. Finally, SRF and actin dynamics are connected via the actin severing protein cofilin and its slingshot phosphatase to modulate neuronal mitochondrial dynamics. In summary, our data suggest that the SRF-cofilin-actin signaling axis modulates neuronal mitochondrial function.
Collapse
|
39
|
Small EM. The actin-MRTF-SRF gene regulatory axis and myofibroblast differentiation. J Cardiovasc Transl Res 2012; 5:794-804. [PMID: 22898751 DOI: 10.1007/s12265-012-9397-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/01/2012] [Indexed: 12/14/2022]
Abstract
Cardiac fibroblasts are responsible for necrotic tissue replacement and scar formation after myocardial infarction (MI) and contribute to remodeling in response to pathological stimuli. This response to insult or injury is largely due to the phenotypic plasticity of fibroblasts. When fibroblasts encounter environmental disturbances, whether biomechanical or humoral, they often transform into smooth muscle-like, contractile cells called "myofibroblasts." The signals that control myofibroblast differentiation include the transforming growth factor (TGF)-β1-Smad pathway and Rho GTPase-dependent actin polymerization. Recent evidence implicates serum response factor (SRF) and the myocardin-related transcription factors (MRTFs) as key mediators of the contractile gene program in response to TGF-β1 or RhoA signaling. This review highlights the function of myofibroblasts in cardiac remodeling and the role of the actin-MRTF-SRF signaling axis in regulating this process.
Collapse
Affiliation(s)
- Eric M Small
- Aab Cardiovascular Research Institute, Department of Medicine, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14642, USA.
| |
Collapse
|
40
|
Takagaki Y, Yamagishi H, Matsuoka R. Factors Involved in Signal Transduction During Vertebrate Myogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:187-272. [DOI: 10.1016/b978-0-12-394307-1.00004-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Nanda V, Miano JM. Leiomodin 1, a new serum response factor-dependent target gene expressed preferentially in differentiated smooth muscle cells. J Biol Chem 2011; 287:2459-67. [PMID: 22157009 DOI: 10.1074/jbc.m111.302224] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Smooth muscle cell (SMC) differentiation is defined largely by a number of cell-restricted genes governed directly by the serum response factor (SRF)/myocardin (MYOCD) transcriptional switch. Here, we describe a new SRF/MYOCD-dependent, SMC-restricted gene known as Leiomodin 1 (Lmod1). Conventional and quantitative RT-PCRs indicate that Lmod1 mRNA expression is enriched in SMC-containing tissues of the mouse, whereas its two paralogs, Lmod2 and Lmod3, exhibit abundant expression in skeletal and cardiac muscle with very low levels in SMC-containing tissues. Western blotting and immunostaining of various adult and embryonic mouse tissues further confirm SMC-specific expression of the LMOD1 protein. Comparative genomic analysis of the human LMOD1 and LMOD2 genes with their respective mouse and rat orthologs shows high conservation between the three exons and several noncoding sequences, including the immediate 5' promoter region. Two conserved CArG boxes are present in both the LMOD1 and LMOD2 promoter regions, although LMOD1 displays much higher promoter activity and is more responsive to SRF/MYOCD stimulation. Gel shift assays demonstrate clear binding between SRF and the two CArG boxes in human LMOD1. Although the CArG boxes in LMOD1 and LMOD2 are similar, only LMOD1 displays SRF or MYOCD-dependent activation. Transgenic mouse studies reveal wild type LMOD1 promoter activity in cardiac and vascular SMC. Such activity is abolished upon mutation of both CArG boxes. Collectively, these data demonstrate that Lmod1 is a new SMC-restricted SRF/MYOCD target gene.
Collapse
Affiliation(s)
- Vivek Nanda
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | |
Collapse
|