1
|
Salmon-Cabrales IS, de la Garza-Kalife DA, García-González G, Estrada-Rodríguez AE, Jiménez-Gutiérrez MA, Santoyo-Suárez MG, Rodríguez-Núñez O, Garza-Treviño EN, Benítez-Chao DF, Padilla-Rivas GR, Islas JF. Exploring the Functionality of the Krüppel-like Factors in Kidney Development, Metabolism, and Diseases. Life (Basel) 2024; 14:1671. [PMID: 39768378 PMCID: PMC11728015 DOI: 10.3390/life14121671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
The kidneys contribute to the overall health of an organism by maintaining systemic homeostasis. This process involves various biological mechanisms, in which the Krüppel-like factors (KLFs), a family of transcription factors, are essential for regulating development, differentiation, proliferation, and cellular apoptosis. They also play a role in the metabolic regulation of essential nutrients, such as glucose and lipids. The dysregulation of these transcription factors is associated with the development of various pathologies, which can ultimately lead to renal fibrosis, severely compromising kidney function. In this context, the present article provides a comprehensive review of the existing literature, offering an enriching analysis of the findings related to the role of KLFs in nephrology, while also highlighting their potential therapeutic role in the treatment of renal diseases.
Collapse
Affiliation(s)
- Itzel S. Salmon-Cabrales
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - David A. de la Garza-Kalife
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Gabriel García-González
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Ana E. Estrada-Rodríguez
- Departmento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico;
| | - Marco Antonio Jiménez-Gutiérrez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Michelle G. Santoyo-Suárez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Oscar Rodríguez-Núñez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Diego F. Benítez-Chao
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Gerardo R. Padilla-Rivas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| | - Jose Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (I.S.S.-C.); (D.A.d.l.G.-K.); (G.G.-G.); (M.A.J.-G.); (M.G.S.-S.); (O.R.-N.); (E.N.G.-T.); (D.F.B.-C.); (G.R.P.-R.)
| |
Collapse
|
2
|
Xu R, Chen Y, Wei S, Chen J. Comprehensive Pan-Cancer Analysis of the Prognostic Role of KLF Transcription Factor 2 (KLF2) in Human Tumors. Onco Targets Ther 2024; 17:887-904. [PMID: 39507409 PMCID: PMC11539754 DOI: 10.2147/ott.s476179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
Background KLF2 is a transcription factor expressed early in mammalian development that plays a role in many processes of development and disease. Recently, increasing studies revealed that KLF2 plays a key role in the occurrence and progression of cancer. Purpose The aim of this study was to explore the role of KLF2 in various tumor types using the Cancer Genome Atlas dataset. Methods Here, we set out to explore the role of KLF2 in 33 tumor types using TCGA (The Cancer Genome Atlas), GEO (Gene Expression Omnibus) dataset, Human Protein Atlas (HPA), UALCAN database, CancerSEA, GSCALite and several bioinformatic tools. Furthermore, we also performed immunohistochemistry and qPCR to further validate the role of KLF2 in multiple cancers and its correlation with prognosis. Results We found that KLF2 was underexpressed in most tumors and generally predicted poor OS in tumor patients. We found that amplification of KLF2 may be a risk factor for patients with OV (Ovarian serous cystadenocarcinoma). We also analyzed the abundance of checkpoints and markers of specific immune subsets including CD8+ T lymphocytes (T cells), CD4+ T cells, macrophages, and endothelial cells that significantly correlated with the expression level of KLF2 in pan-carcinoma tissues. In some cancers, KLF2 expression levels are positively correlated with gene promoter DNA methylation and drug sensitivity. In addition, we found that KLF2 is involved in single-cell level cell invasion in some cancers. In addition, KLF2 is co-expressed with several intracellular signal transduction genes involved in immune system processes. Immunohistochemistry and qPCR confirmed the low expression of KLF2 in STAD (stomach adenocarcinoma) and renal cancer. Conclusion Our pan-cancer analysis provides a comprehensive overview of the oncogenic roles of KLF2 in multiple human cancers and can be regarded as a potential prognostic marker and a novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Rong Xu
- Department of Dermatology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yuhan Chen
- Department of Pathology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Shicai Wei
- Department of Dermatology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jun Chen
- Department of Dermatology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
3
|
Kim RT, Whited JL. Putative epithelial-mesenchymal transitions during salamander limb regeneration: Current perspectives and future investigations. Ann N Y Acad Sci 2024; 1540:89-103. [PMID: 39269330 PMCID: PMC11471381 DOI: 10.1111/nyas.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Previous studies have implicated epithelial-mesenchymal transition (EMT) in salamander limb regeneration. In this review, we describe putative roles for EMT during each stage of limb regeneration in axolotls and other salamanders. We hypothesize that EMT and EMT-like gene expression programs may regulate three main cellular processes during limb regeneration: (1) keratinocyte migration during wound closure; (2) transient invasion of the stump by epithelial cells undergoing EMT; and (3) use of EMT-like programs by non-epithelial blastemal progenitor cells to escape the confines of their niches. Finally, we propose nontraditional roles for EMT during limb regeneration that warrant further investigation, including alternative EMT regulators, stem cell activation, and fibrosis induced by aberrant EMT.
Collapse
Affiliation(s)
- Ryan T Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
da Silva AR, Gunawan F, Boezio GLM, Faure E, Théron A, Avierinos JF, Lim S, Jha SG, Ramadass R, Guenther S, Looso M, Zaffran S, Juan T, Stainier DYR. egr3 is a mechanosensitive transcription factor gene required for cardiac valve morphogenesis. SCIENCE ADVANCES 2024; 10:eadl0633. [PMID: 38748804 PMCID: PMC11095463 DOI: 10.1126/sciadv.adl0633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024]
Abstract
Biomechanical forces, and their molecular transducers, including key mechanosensitive transcription factor genes, such as KLF2, are required for cardiac valve morphogenesis. However, klf2 mutants fail to completely recapitulate the valveless phenotype observed under no-flow conditions. Here, we identify the transcription factor EGR3 as a conserved biomechanical force transducer critical for cardiac valve formation. We first show that egr3 null zebrafish display a complete and highly penetrant loss of valve leaflets, leading to severe blood regurgitation. Using tissue-specific loss- and gain-of-function tools, we find that during cardiac valve formation, Egr3 functions cell-autonomously in endothelial cells, and identify one of its effectors, the nuclear receptor Nr4a2b. We further find that mechanical forces up-regulate egr3/EGR3 expression in the developing zebrafish heart and in porcine valvular endothelial cells, as well as during human aortic valve remodeling. Altogether, these findings reveal that EGR3 is necessary to transduce the biomechanical cues required for zebrafish cardiac valve morphogenesis, and potentially for pathological aortic valve remodeling in humans.
Collapse
Affiliation(s)
- Agatha Ribeiro da Silva
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Giulia L. M. Boezio
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Emilie Faure
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Alexis Théron
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Chirurgie Cardiaque, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - Jean-François Avierinos
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Cardiologie, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - SoEun Lim
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Shivam Govind Jha
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Radhan Ramadass
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Stefan Guenther
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stéphane Zaffran
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Thomas Juan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| |
Collapse
|
5
|
García-Loredo JA, Santoyo-Suarez MG, Rodríguez-Nuñez O, Benitez Chao DF, Garza-Treviño EN, Zapata-Morin PA, Padilla-Rivas GR, Islas JF. Is the Cis-Element CACCC-Box a Master Regulatory Element during Cardiovascular Disease? A Bioinformatics Approach from the Perspective of the Krüppel-like Family of Transcription Factors. Life (Basel) 2024; 14:493. [PMID: 38672763 PMCID: PMC11051458 DOI: 10.3390/life14040493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The CACCC-box motif emerges as a pivotal cis-regulatory element implicated in diverse developmental processes and diseases, particularly cardiovascular diseases (CVDs). This study centers on the intricate interplay between the CACCC-box and its binding proteins such as: the Krüppel-Like Family (KLF) of transcription factors as primary effectors in the context of CVDs. Our analysis was through a bioinformatics approach, which revealed significant transcriptional activity among KLF subgroup 2, exhibiting the highest number of interactions focusing on the established roles: pluripotency, cancer, and cardiovascular development and diseases. Our analysis reveals KLF's interactions with GATA4, MEF2C, NKX2.5 and other ~90 potential genes that participate in the regulation of the hypertrophic environment (or CVDs' Environment). Also, the GO analysis showed that genes containing the motif CACCC were enriched for multiple CVDs; in combination with STRING analysis, these results pointed to a link between KLFs and these diseases. The analysis further identifies other potential CACCC-box binding factors, such as SP family members, WT1, VEZF1, and -SALL4, which are implicated in cardiac contraction, remodeling, and inflammation processes.
Collapse
Affiliation(s)
- Juan Andrés García-Loredo
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
- Laboratorio de Micología y Fitopatología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo León, Mexico;
| | - Michelle G. Santoyo-Suarez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Oscar Rodríguez-Nuñez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Diego Francisco Benitez Chao
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Patricio Adrián Zapata-Morin
- Laboratorio de Micología y Fitopatología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo León, Mexico;
| | - Gerardo R. Padilla-Rivas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Jose Francisco Islas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| |
Collapse
|
6
|
Tsitsikov EN, Phan KP, Liu Y, Tsytsykova AV, Kinter M, Selland L, Garman L, Griffin C, Dunn IF. TRAF7 is an essential regulator of blood vessel integrity during mouse embryonic and neonatal development. iScience 2023; 26:107474. [PMID: 37583551 PMCID: PMC10424150 DOI: 10.1016/j.isci.2023.107474] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/19/2023] [Accepted: 07/21/2023] [Indexed: 08/17/2023] Open
Abstract
Targeted deletion of TRAF7 revealed that it is a crucial part of shear stress-responsive MEKK3-MEK5-ERK5 signaling pathway induced in endothelial cells by blood flow. Similar to Mekk3-, Mek5- or Erk5-deficient mice, Traf7-deficient embryos died in utero around midgestation due to impaired endothelium integrity. They displayed significantly lower expression of transcription factor Klf2, an essential regulator of vascular hemodynamic forces downstream of the MEKK3-MEK-ERK5 signaling pathway. In addition, deletion of Traf7 in endothelial cells of postnatal mice was associated with severe cerebral hemorrhage. Here, we show that besides MEKK3 and MEK5, TRAF7 associates with a planar cell polarity protein SCRIB. SCRIB binds with an N-terminal region of TRAF7, while MEKK3 associates with the C-terminal WD40 domain. Downregulation of TRAF7 as well as SCRIB inhibited fluid shear stress-induced phosphorylation of ERK5 in cultured endothelial cells. These findings suggest that TRAF7 and SCRIB may comprise an upstream part of the MEKK3-MEK5-ERK5 signaling pathway.
Collapse
Affiliation(s)
- Erdyni N. Tsitsikov
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Khanh P. Phan
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yufeng Liu
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alla V. Tsytsykova
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mike Kinter
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Lauren Selland
- Histology, Immunohistochemistry, Microscopy Core-COBRE Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lori Garman
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Courtney Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Ian F. Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
7
|
George RM, Firulli BA, Podicheti R, Rusch DB, Mannion BJ, Pennacchio LA, Osterwalder M, Firulli AB. Single cell evaluation of endocardial Hand2 gene regulatory networks reveals HAND2-dependent pathways that impact cardiac morphogenesis. Development 2023; 150:dev201341. [PMID: 36620995 PMCID: PMC10110492 DOI: 10.1242/dev.201341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
The transcription factor HAND2 plays essential roles during cardiogenesis. Hand2 endocardial deletion (H2CKO) results in tricuspid atresia or double inlet left ventricle with accompanying intraventricular septum defects, hypo-trabeculated ventricles and an increased density of coronary lumens. To understand the regulatory mechanisms of these phenotypes, single cell transcriptome analysis of mouse E11.5 H2CKO hearts was performed revealing a number of disrupted endocardial regulatory pathways. Using HAND2 DNA occupancy data, we identify several HAND2-dependent enhancers, including two endothelial enhancers for the shear-stress master regulator KLF2. A 1.8 kb enhancer located 50 kb upstream of the Klf2 TSS imparts specific endothelial/endocardial expression within the vasculature and endocardium. This enhancer is HAND2-dependent for ventricular endocardium expression but HAND2-independent for Klf2 vascular and valve expression. Deletion of this Klf2 enhancer results in reduced Klf2 expression within ventricular endocardium. These data reveal that HAND2 functions within endocardial gene regulatory networks including shear-stress response.
Collapse
Affiliation(s)
- Rajani M. George
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Beth A. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Len A. Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern 3008, Switzerland
- Department of Cardiology, Bern University Hospital, Bern 3010, Switzerland
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN 46202, USA
| |
Collapse
|
8
|
Niu Q, Gao J, Wang L, Liu J, Zhang L. Regulation of differentiation and generation of osteoclasts in rheumatoid arthritis. Front Immunol 2022; 13:1034050. [PMID: 36466887 PMCID: PMC9716075 DOI: 10.3389/fimmu.2022.1034050] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 09/25/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA), which affects nearly 1% of the world's population, is a debilitating autoimmune disease. Bone erosion caused by periarticular osteopenia and synovial pannus formation is the most destructive pathological changes of RA, also leads to joint deformity and loss of function,and ultimately affects the quality of life of patients. Osteoclasts (OCs) are the only known bone resorption cells and their abnormal differentiation and production play an important role in the occurrence and development of RA bone destruction; this remains the main culprit behind RA. METHOD Based on the latest published literature and research progress at home and abroad, this paper reviews the abnormal regulation mechanism of OC generation and differentiation in RA and the possible targeted therapy. RESULT OC-mediated bone destruction is achieved through the regulation of a variety of cytokines and cell-to-cell interactions, including gene transcription, epigenetics and environmental factors. At present, most methods for the treatment of RA are based on the regulation of inflammation, the inhibition of bone injury and joint deformities remains unexplored. DISCUSSION This article will review the mechanism of abnormal differentiation of OC in RA, and summarise the current treatment oftargeting cytokines in the process of OC generation and differentiation to reduce bone destruction in patients with RA, which isexpected to become a valuable treatment choice to inhibit bone destruction in RA.
Collapse
Affiliation(s)
- Qing Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiaxi Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
9
|
Feulner L, van Vliet PP, Puceat M, Andelfinger G. Endocardial Regulation of Cardiac Development. J Cardiovasc Dev Dis 2022; 9:jcdd9050122. [PMID: 35621833 PMCID: PMC9144171 DOI: 10.3390/jcdd9050122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 01/16/2023] Open
Abstract
The endocardium is a specialized form of endothelium that lines the inner side of the heart chambers and plays a crucial role in cardiac development. While comparatively less studied than other cardiac cell types, much progress has been made in understanding the regulation of and by the endocardium over the past two decades. In this review, we will summarize what is currently known regarding endocardial origin and development, the relationship between endocardium and other cardiac cell types, and the various lineages that endocardial cells derive from and contribute to. These processes are driven by key molecular mechanisms such as Notch and BMP signaling. These pathways in particular have been well studied, but other signaling pathways and mechanical cues also play important roles. Finally, we will touch on the contribution of stem cell modeling in combination with single cell sequencing and its potential translational impact for congenital heart defects such as bicuspid aortic valves and hypoplastic left heart syndrome. The detailed understanding of cellular and molecular processes in the endocardium will be vital to further develop representative stem cell-derived models for disease modeling and regenerative medicine in the future.
Collapse
Affiliation(s)
- Lara Feulner
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- Department of Molecular Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Patrick Piet van Vliet
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- LIA (International Associated Laboratory) CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- LIA (International Associated Laboratory) INSERM, 13885 Marseille, France
| | - Michel Puceat
- LIA (International Associated Laboratory) CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- LIA (International Associated Laboratory) INSERM, 13885 Marseille, France
- INSERM U-1251, Marseille Medical Genetics, Aix-Marseille University, 13885 Marseille, France
| | - Gregor Andelfinger
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Biochemistry, University of Montreal, Montreal, QC H3T 1J4, Canada
- Correspondence:
| |
Collapse
|
10
|
Endothelial MEKK3-KLF2/4 signaling integrates inflammatory and hemodynamic signals during definitive hematopoiesis. Blood 2022; 139:2942-2957. [PMID: 35245372 PMCID: PMC9101247 DOI: 10.1182/blood.2021013934] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
The hematopoietic stem cells (HSCs) that produce blood for the lifetime of an animal arise from RUNX1+ hemogenic endothelial cells (HECs) in the embryonic vasculature through a process of endothelial-to-hematopoietic transition (EHT). Studies have identified inflammatory mediators and fluid shear forces as critical environmental stimuli for EHT, raising the question of how such diverse inputs are integrated to drive HEC specification. Endothelial cell MEKK3-KLF2/4 signaling can be activated by both fluid shear forces and inflammatory mediators, and plays roles in cardiovascular development and disease that have been linked to both stimuli. Here we demonstrate that MEKK3 and KLF2/4 are required in endothelial cells for the specification of RUNX1+ HECs in both the yolk sac and dorsal aorta of the mouse embryo and for their transition to intra-aortic hematopoietic cluster cells (IAHCs). The inflammatory mediators lipopolysaccharide and interferon gamma increase RUNX1+ HECs in an MEKK3-dependent manner. Maternal administration of catecholamines that stimulate embryo cardiac function and accelerate yolk sac vascular remodeling increases EHT by wild-type but not MEKK3-deficient endothelium. These findings identify MEKK-KLF2/4 signaling as an essential pathway for EHT and provide a molecular basis for the integration of diverse environmental inputs, such as inflammatory mediators and hemodynamic forces, during definitive hematopoiesis.
Collapse
|
11
|
Jiang S, Luo M, Bai X, Nie P, Zhu Y, Cai H, Li B, Luo P. Cellular crosstalk of glomerular endothelial cells and podocytes in diabetic kidney disease. J Cell Commun Signal 2022; 16:313-331. [PMID: 35041192 DOI: 10.1007/s12079-021-00664-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious microvascular complication of diabetes and is the leading cause of end-stage renal disease (ESRD). Persistent proteinuria is an important feature of DKD, which is caused by the destruction of the glomerular filtration barrier (GFB). Glomerular endothelial cells (GECs) and podocytes are important components of the GFB, and their damage can be observed in the early stages of DKD. Recently, studies have found that crosstalk between cells directly affects DKD progression, which has prospective research significance. However, the pathways involved are complex and largely unexplored. Here, we review the literature on cellular crosstalk of GECs and podocytes in the context of DKD, and highlight specific gaps in the field to propose future research directions. Elucidating the intricates of such complex processes will help to further understand the pathogenesis of DKD and develop better prevention and treatment options.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Hangxi Cai
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
12
|
Hypoplastic left heart syndrome (HLHS): molecular pathogenesis and emerging drug targets for cardiac repair and regeneration. Expert Opin Ther Targets 2021; 25:621-632. [PMID: 34488532 DOI: 10.1080/14728222.2021.1978069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Hypoplastic left heart syndrome (HLHS) is a severe developmental defect characterized by the underdevelopment of the left ventricle along with aortic and valvular defects. Multiple palliative surgeries are required for survival. Emerging studies have identified potential mechanisms for the disease onset, including genetic and hemodynamic causes. Genetic variants associated with HLHS include transcription factors, chromatin remodelers, structural proteins, and signaling proteins necessary for normal heart development. Nonetheless, current therapies are being tested clinically and have shown promising results at improving cardiac function in patients who have undergone palliative surgeries. AREAS COVERED We searched PubMed and clinicaltrials.gov to review most of the mechanistic research and clinical trials involving HLHS. This review discusses the anatomy and pathology of HLHS hearts. We highlight some of the identified genetic variants that underly the molecular pathogenesis of HLHS. Additionally, we discuss some of the emerging therapies and their limitations for HLHS. EXPERT OPINION While HLHS etiology is largely obscure, palliative therapies remain the most viable option for the patients. It is necessary to generate animal and stem cell models to understand the underlying genetic causes directly leading to HLHS and facilitate the use of gene-based therapies to improve cardiac development and regeneration.
Collapse
|
13
|
Fontana F, Haack T, Reichenbach M, Knaus P, Puceat M, Abdelilah-Seyfried S. Antagonistic Activities of Vegfr3/Flt4 and Notch1b Fine-tune Mechanosensitive Signaling during Zebrafish Cardiac Valvulogenesis. Cell Rep 2021; 32:107883. [PMID: 32668254 DOI: 10.1016/j.celrep.2020.107883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022] Open
Abstract
The formation of cardiac valves depends on mechanical forces exerted by blood flow. Endocardial cells lining the interior of the heart are sensitive to these stimuli and respond by rearranging into luminal cells subjected to shear stress and abluminal cells not exposed to it. The mechanisms by which endocardial cells sense these dynamic biomechanical stimuli and how they evoke different cellular responses are largely unknown. Here, we show that blood flow activates two parallel mechanosensitive pathways, one mediated by Notch and the other by Klf2a. Both pathways negatively regulate the angiogenesis receptor Vegfr3/Flt4, which becomes restricted to abluminal endocardial cells. Its loss disrupts valve morphogenesis and results in the occurrence of Notch signaling within abluminal endocardial cells. Our work explains how antagonistic activities by Vegfr3/Flt4 on the abluminal side and by Notch on the luminal side shape cardiac valve leaflets by triggering unique differences in the fates of endocardial cells.
Collapse
Affiliation(s)
- Federica Fontana
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| | - Timm Haack
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany
| | - Maria Reichenbach
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Petra Knaus
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Michel Puceat
- INSERM U-1251, MMG, Aix-Marseille University, 13885 Marseille, France
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany.
| |
Collapse
|
14
|
Abstract
The developing heart is formed of two tissue layers separated by an extracellular matrix (ECM) that provides chemical and physical signals to cardiac cells. While deposition of specific ECM components creates matrix diversity, the cardiac ECM is also dynamic, with modification and degradation playing important roles in ECM maturation and function. In this Review, we discuss the spatiotemporal changes in ECM composition during cardiac development that support distinct aspects of heart morphogenesis. We highlight conserved requirements for specific ECM components in human cardiac development, and discuss emerging evidence of a central role for the ECM in promoting heart regeneration.
Collapse
Affiliation(s)
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
15
|
Abstract
Cardiac development is a complex developmental process that is initiated soon after gastrulation, as two sets of precardiac mesodermal precursors are symmetrically located and subsequently fused at the embryonic midline forming the cardiac straight tube. Thereafter, the cardiac straight tube invariably bends to the right, configuring the first sign of morphological left–right asymmetry and soon thereafter the atrial and ventricular chambers are formed, expanded and progressively septated. As a consequence of all these morphogenetic processes, the fetal heart acquired a four-chambered structure having distinct inlet and outlet connections and a specialized conduction system capable of directing the electrical impulse within the fully formed heart. Over the last decades, our understanding of the morphogenetic, cellular, and molecular pathways involved in cardiac development has exponentially grown. Multiples aspects of the initial discoveries during heart formation has served as guiding tools to understand the etiology of cardiac congenital anomalies and adult cardiac pathology, as well as to enlighten novels approaches to heal the damaged heart. In this review we provide an overview of the complex cellular and molecular pathways driving heart morphogenesis and how those discoveries have provided new roads into the genetic, clinical and therapeutic management of the diseased hearts.
Collapse
|
16
|
Liu X, Chen W, Li W, Li Y, Priest JR, Zhou B, Wang J, Zhou Z. Single-Cell RNA-Seq of the Developing Cardiac Outflow Tract Reveals Convergent Development of the Vascular Smooth Muscle Cells. Cell Rep 2020; 28:1346-1361.e4. [PMID: 31365875 DOI: 10.1016/j.celrep.2019.06.092] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac outflow tract (OFT) is a major hotspot for congenital heart diseases. A thorough understanding of the cellular diversity, transitions, and regulatory networks of normal OFT development is essential to decipher the etiology of OFT malformations. We performed single-cell transcriptomic sequencing of 55,611 mouse OFT cells from three developmental stages that generally correspond to the early, middle, and late stages of OFT remodeling and septation. Known cellular transitions, such as endothelial-to-mesenchymal transition, have been recapitulated. In particular, we identified convergent development of the vascular smooth muscle cell (VSMC) lineage where intermediate cell subpopulations were found to be involved in either myocardial-to-VSMC trans-differentiation or mesenchymal-to-VSMC transition. Finally, we uncovered transcriptional regulators potentially governing cellular transitions. Our study provides a single-cell reference map of cell states for normal OFT development and paves the way for further studies of the etiology of OFT malformations at the single-cell level.
Collapse
Affiliation(s)
- Xuanyu Liu
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wen Chen
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wenke Li
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - James R Priest
- Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University. School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Jikui Wang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University. Xinxiang 453003, China.
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| |
Collapse
|
17
|
Zimmer BM, Barycki JJ, Simpson MA. Integration of Sugar Metabolism and Proteoglycan Synthesis by UDP-glucose Dehydrogenase. J Histochem Cytochem 2020; 69:13-23. [PMID: 32749901 DOI: 10.1369/0022155420947500] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Regulation of proteoglycan and glycosaminoglycan synthesis is critical throughout development, and to maintain normal adult functions in wound healing and the immune system, among others. It has become increasingly clear that these processes are also under tight metabolic control and that availability of carbohydrate and amino acid metabolite precursors has a role in the control of proteoglycan and glycosaminoglycan turnover. The enzyme uridine diphosphate (UDP)-glucose dehydrogenase (UGDH) produces UDP-glucuronate, an essential precursor for new glycosaminoglycan synthesis that is tightly controlled at multiple levels. Here, we review the cellular mechanisms that regulate UGDH expression, discuss the structural features of the enzyme, and use the structures to provide a context for recent studies that link post-translational modifications and allosteric modulators of UGDH to its function in downstream pathways.
Collapse
Affiliation(s)
- Brenna M Zimmer
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina
| | - Joseph J Barycki
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina
| | - Melanie A Simpson
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
18
|
Rolph D, Das H. Transcriptional Regulation of Osteoclastogenesis: The Emerging Role of KLF2. Front Immunol 2020; 11:937. [PMID: 32477372 PMCID: PMC7237574 DOI: 10.3389/fimmu.2020.00937] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of osteoclastic differentiation and its activity is a hallmark of various musculoskeletal disease states. In this review, the complex molecular factors underlying osteoclastic differentiation and function are evaluated. The emerging role of KLF2 in regulation of osteoclastic differentiation is examined, specifically in the context of rheumatoid arthritis in which it has been most extensively studied among the musculoskeletal diseases. The therapies that exist to manage diseases associated with osteoclastogenesis are numerous and diverse. They are varied in their mechanisms of action and in the outcomes they produce. For this review, therapies targeting osteoclasts will be emphasized, though it should be noted that many therapies exist which bolster the action of osteoblasts. A new targeted molecular approach is under investigation for the future potential therapeutic development of rheumatoid arthritis.
Collapse
Affiliation(s)
- Daniela Rolph
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| |
Collapse
|
19
|
Chiu YL, Tsai WC, Wu CH, Wu CH, Cheng CC, Lin WS, Tsai TN, Wu LS. Ginkgo biloba Induces Thrombomodulin Expression and Tissue-Type Plasminogen Activator Secretion via the Activation of Krüppel-Like Factor 2 within Endothelial Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:357-372. [PMID: 32108493 DOI: 10.1142/s0192415x20500184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effects of thrombo-prevention, such as antiplatelet and anticoagulant activity, have been reported with the usage of Ginkgo biloba extract (GbE); however, the detailed mechanism has not yet been fully investigated, especially the role of Krüppel-like factor 2 (KLF2). This study aimed to investigate whether GbE can activate KLF2 and then induce thrombomodulin (TM) and tissue-type plasminogen activator (t-PA) secretion to enhance the effects of thrombo-prevention. Different concentrations of GbE were incubated with human umbilical vein endothelial cells (HUVECs) to evaluate its effect on endothelial cells. We found that KLF2 expression is correlated to the risk of atherosclerosis and venous thromboembolism in clinical practice. In the HUVEC cell model, GbE stimulated the expression of KLF2 in a dose-dependent manner. Moreover, TM and t-PA secretion increased when the cells were cultured with GbE. Both the expressions and activities of TM and t-PA in the GbE-treated cells declined after KLF2 was blocked by shKLF2. In sum, with GbE treatment, KLF2 expression in human endothelial cells was significantly activated, which in turn induced an increase in the protein expression and activity of TM and t-PA. After shRNA inhibited the KLF2 expression, GbE stopped inducing the expression and activity of TM and t-PA. These findings suggest that GbE exerts an antithrombotic effect on endothelial cells by increasing the TM expression and t-PA secretion; further, KLF2 is a key factor in this mechanism.
Collapse
Affiliation(s)
- Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Wei-Che Tsai
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Chih-Hsien Wu
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Chun-Hsien Wu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Cheng-Chung Cheng
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Wei-Shing Lin
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Tsung-Neng Tsai
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan, R.O.C.,Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Lian-Shan Wu
- Division of Cardiology, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien County 971, Taiwan, R.O.C
| |
Collapse
|
20
|
Qu X, Violette K, Sewell-Loftin MK, Soslow J, Saint-Jean L, Hinton RB, Merryman WD, Baldwin HS. Loss of flow responsive Tie1 results in Impaired
Aortic valve remodeling. Dev Biol 2019; 455:73-84. [PMID: 31319059 DOI: 10.1016/j.ydbio.2019.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 10/26/2022]
Abstract
The mechanisms regulating endothelial cell response to hemodynamic forces required for heart valve development, especially valve remodeling, remain elusive. Tie1, an endothelial specific receptor tyrosine kinase, is up-regulated by oscillating shear stress and is required for lymphatic valve development. In this study, we demonstrate that valvular endothelial Tie1 is differentially expressed in a dynamic pattern predicted by disturbed flow during valve remodeling. Following valvular endocardial specific deletion of Tie1 in mice, we observed enlarged aortic valve leaflets, decreased valve stiffness and valvular insufficiency. Valve abnormalities were only detected in late gestation and early postnatal mutant animals and worsened with age. The mutant mice developed perturbed extracellular matrix (ECM) deposition and remodeling characterized by increased glycosaminoglycan and decreased collagen content, as well as increased valve interstitial cell expression of Sox9, a transcription factor essential for normal ECM maturation during heart valve development. This study provides the first evidence that Tie1 is involved in modulation of late valve remodeling and suggests that an important Tie1-Sox9 signaling axis exists through which disturbed flows are converted by endocardial cells to paracrine Sox9 signals to modulate normal matrix remodeling of the aortic valve.
Collapse
Affiliation(s)
- Xianghu Qu
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - Kate Violette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - M K Sewell-Loftin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonathan Soslow
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - LeShana Saint-Jean
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert B Hinton
- Division of Cardiology, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - H Scott Baldwin
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
21
|
Poelmann RE, Gittenberger-de Groot AC. Hemodynamics in Cardiac Development. J Cardiovasc Dev Dis 2018; 5:jcdd5040054. [PMID: 30404214 PMCID: PMC6306789 DOI: 10.3390/jcdd5040054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/03/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
The beating heart is subject to intrinsic mechanical factors, exerted by contraction of the myocardium (stretch and strain) and fluid forces of the enclosed blood (wall shear stress). The earliest contractions of the heart occur already in the 10-somite stage in the tubular as yet unsegmented heart. With development, the looping heart becomes asymmetric providing varying diameters and curvatures resulting in unequal flow profiles. These flow profiles exert various wall shear stresses and as a consequence different expression patterns of shear responsive genes. In this paper we investigate the morphological alterations of the heart after changing the blood flow by ligation of the right vitelline vein in a model chicken embryo and analyze the extended expression in the endocardial cushions of the shear responsive gene Tgfbeta receptor III. A major phenomenon is the diminished endocardial-mesenchymal transition resulting in hypoplastic (even absence of) atrioventricular and outflow tract endocardial cushions, which might be lethal in early phases. The surviving embryos exhibit several cardiac malformations including ventricular septal defects and malformed semilunar valves related to abnormal development of the aortopulmonary septal complex and the enclosed neural crest cells. We discuss the results in the light of the interactions between several shear stress responsive signaling pathways including an extended review of the involved Vegf, Notch, Pdgf, Klf2, eNos, Endothelin and Tgfβ/Bmp/Smad networks.
Collapse
Affiliation(s)
- Robert E Poelmann
- Department of Animal Sciences and Health, Institute of Biology, Sylvius Laboratory, University of Leiden, Sylviusweg 72, 2333BE Leiden, The Netherlands.
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 20, 2300RC Leiden, The Netherlands.
| | | |
Collapse
|
22
|
Geng X, Cha B, Mahamud MR, Srinivasan RS. Intraluminal valves: development, function and disease. Dis Model Mech 2018; 10:1273-1287. [PMID: 29125824 PMCID: PMC5719258 DOI: 10.1242/dmm.030825] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The circulatory system consists of the heart, blood vessels and lymphatic vessels, which function in parallel to provide nutrients and remove waste from the body. Vascular function depends on valves, which regulate unidirectional fluid flow against gravitational and pressure gradients. Severe valve disorders can cause mortality and some are associated with severe morbidity. Although cardiac valve defects can be treated by valve replacement surgery, no treatment is currently available for valve disorders of the veins and lymphatics. Thus, a better understanding of valves, their development and the progression of valve disease is warranted. In the past decade, molecules that are important for vascular function in humans have been identified, with mouse studies also providing new insights into valve formation and function. Intriguing similarities have recently emerged between the different types of valves concerning their molecular identity, architecture and development. Shear stress generated by fluid flow has also been shown to regulate endothelial cell identity in valves. Here, we review our current understanding of valve development with an emphasis on its mechanobiology and significance to human health, and highlight unanswered questions and translational opportunities.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Md Riaj Mahamud
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
23
|
Bioinformatics analysis of non-synonymous variants in the KLF genes related to cardiac diseases. Gene 2018; 650:68-76. [DOI: 10.1016/j.gene.2018.01.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/23/2017] [Accepted: 01/25/2018] [Indexed: 12/25/2022]
|
24
|
Goddard LM, Duchemin AL, Ramalingan H, Wu B, Chen M, Bamezai S, Yang J, Li L, Morley MP, Wang T, Scherrer-Crosbie M, Frank DB, Engleka KA, Jameson SC, Morrisey EE, Carroll TJ, Zhou B, Vermot J, Kahn ML. Hemodynamic Forces Sculpt Developing Heart Valves through a KLF2-WNT9B Paracrine Signaling Axis. Dev Cell 2017; 43:274-289.e5. [PMID: 29056552 DOI: 10.1016/j.devcel.2017.09.023] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 08/01/2017] [Accepted: 09/25/2017] [Indexed: 01/08/2023]
Abstract
Hemodynamic forces play an essential epigenetic role in heart valve development, but how they do so is not known. Here, we show that the shear-responsive transcription factor KLF2 is required in endocardial cells to regulate the mesenchymal cell responses that remodel cardiac cushions to mature valves. Endocardial Klf2 deficiency results in defective valve formation associated with loss of Wnt9b expression and reduced canonical WNT signaling in neighboring mesenchymal cells, a phenotype reproduced by endocardial-specific loss of Wnt9b. Studies in zebrafish embryos reveal that wnt9b expression is similarly restricted to the endocardial cells overlying the developing heart valves and is dependent upon both hemodynamic shear forces and klf2a expression. These studies identify KLF2-WNT9B signaling as a conserved molecular mechanism by which fluid forces sensed by endothelial cells direct the complex cellular process of heart valve development and suggest that congenital valve defects may arise due to subtle defects in this mechanotransduction pathway.
Collapse
Affiliation(s)
- Lauren M Goddard
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Anne-Laure Duchemin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France; Université de Strasbourg, Illkirch 67404, France
| | - Harini Ramalingan
- Department of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Bingruo Wu
- Department of Genetics, Pediatric, and Medicine (Cardiology) and Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine of Yeshiva University, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Mei Chen
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Sharika Bamezai
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Li Li
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Tao Wang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Marielle Scherrer-Crosbie
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - David B Frank
- Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kurt A Engleka
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Edward E Morrisey
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Thomas J Carroll
- Department of Internal Medicine (Nephrology) and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Bin Zhou
- Department of Genetics, Pediatric, and Medicine (Cardiology) and Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine of Yeshiva University, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France; Université de Strasbourg, Illkirch 67404, France
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Bialkowska AB, Yang VW, Mallipattu SK. Krüppel-like factors in mammalian stem cells and development. Development 2017; 144:737-754. [PMID: 28246209 DOI: 10.1242/dev.145441] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors that are found in many species. Recent studies have shown that KLFs play a fundamental role in regulating diverse biological processes such as cell proliferation, differentiation, development and regeneration. Of note, several KLFs are also crucial for maintaining pluripotency and, hence, have been linked to reprogramming and regenerative medicine approaches. Here, we review the crucial functions of KLFs in mammalian embryogenesis, stem cell biology and regeneration, as revealed by studies of animal models. We also highlight how KLFs have been implicated in human diseases and outline potential avenues for future research.
Collapse
Affiliation(s)
- Agnieszka B Bialkowska
- Division of Gastroenterology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| | - Vincent W Yang
- Division of Gastroenterology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA.,Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| |
Collapse
|
26
|
Hill JT, Demarest B, Gorsi B, Smith M, Yost HJ. Heart morphogenesis gene regulatory networks revealed by temporal expression analysis. Development 2017; 144:3487-3498. [PMID: 28807900 DOI: 10.1242/dev.154146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
During embryogenesis the heart forms as a linear tube that then undergoes multiple simultaneous morphogenetic events to obtain its mature shape. To understand the gene regulatory networks (GRNs) driving this phase of heart development, during which many congenital heart disease malformations likely arise, we conducted an RNA-seq timecourse in zebrafish from 30 hpf to 72 hpf and identified 5861 genes with altered expression. We clustered the genes by temporal expression pattern, identified transcription factor binding motifs enriched in each cluster, and generated a model GRN for the major gene batteries in heart morphogenesis. This approach predicted hundreds of regulatory interactions and found batteries enriched in specific cell and tissue types, indicating that the approach can be used to narrow the search for novel genetic markers and regulatory interactions. Subsequent analyses confirmed the GRN using two mutants, Tbx5 and nkx2-5, and identified sets of duplicated zebrafish genes that do not show temporal subfunctionalization. This dataset provides an essential resource for future studies on the genetic/epigenetic pathways implicated in congenital heart defects and the mechanisms of cardiac transcriptional regulation.
Collapse
Affiliation(s)
- Jonathon T Hill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA .,Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Bradley Demarest
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Bushra Gorsi
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Megan Smith
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - H Joseph Yost
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
27
|
Haack T, Abdelilah-Seyfried S. The force within: endocardial development, mechanotransduction and signalling during cardiac morphogenesis. Development 2016; 143:373-86. [PMID: 26839341 DOI: 10.1242/dev.131425] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.
Collapse
Affiliation(s)
- Timm Haack
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, Potsdam D-14476, Germany
| |
Collapse
|
28
|
Steed E, Faggianelli N, Roth S, Ramspacher C, Concordet JP, Vermot J. klf2a couples mechanotransduction and zebrafish valve morphogenesis through fibronectin synthesis. Nat Commun 2016; 7:11646. [PMID: 27221222 PMCID: PMC4894956 DOI: 10.1038/ncomms11646] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 04/15/2016] [Indexed: 12/23/2022] Open
Abstract
The heartbeat and blood flow signal to endocardial cell progenitors through mechanosensitive proteins that modulate the genetic program controlling heart valve morphogenesis. To date, the mechanism by which mechanical forces coordinate tissue morphogenesis is poorly understood. Here we use high-resolution imaging to uncover the coordinated cell behaviours leading to heart valve formation. We find that heart valves originate from progenitors located in the ventricle and atrium that generate the valve leaflets through a coordinated set of endocardial tissue movements. Gene profiling analyses and live imaging reveal that this reorganization is dependent on extracellular matrix proteins, in particular on the expression of fibronectin1b. We show that blood flow and klf2a, a major endocardial flow-responsive gene, control these cell behaviours and fibronectin1b synthesis. Our results uncover a unique multicellular layering process leading to leaflet formation and demonstrate that endocardial mechanotransduction and valve morphogenesis are coupled via cellular rearrangements mediated by fibronectin synthesis.
Collapse
Affiliation(s)
- Emily Steed
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Nathalie Faggianelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Stéphane Roth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Caroline Ramspacher
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Jean-Paul Concordet
- Muséum National d'Histoire Naturelle, 75231 Paris Cedex 05, France
- CNRS UMR 7196, 75231 Paris Cedex 05, France
- INSERM U1154, 75231 Paris Cedex 05, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| |
Collapse
|
29
|
Andrés-Delgado L, Mercader N. Interplay between cardiac function and heart development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1707-16. [PMID: 26952935 PMCID: PMC4906158 DOI: 10.1016/j.bbamcr.2016.03.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 12/24/2022]
Abstract
Mechanotransduction refers to the conversion of mechanical forces into biochemical or electrical signals that initiate structural and functional remodeling in cells and tissues. The heart is a kinetic organ whose form changes considerably during development and disease. This requires cardiomyocytes to be mechanically durable and able to mount coordinated responses to a variety of environmental signals on different time scales, including cardiac pressure loading and electrical and hemodynamic forces. During physiological growth, myocytes, endocardial and epicardial cells have to adaptively remodel to these mechanical forces. Here we review some of the recent advances in the understanding of how mechanical forces influence cardiac development, with a focus on fluid flow forces. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Laura Andrés-Delgado
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Nadia Mercader
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain; Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
30
|
Menon V, Eberth JF, Goodwin RL, Potts JD. Altered Hemodynamics in the Embryonic Heart Affects Outflow Valve Development. J Cardiovasc Dev Dis 2015; 2:108-124. [PMID: 26878022 PMCID: PMC4751060 DOI: 10.3390/jcdd2020108] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cardiac valve structure and function are primarily determined during early development. Consequently, abnormally-formed heart valves are the most common type of congenital heart defects. Several adult valve diseases can be backtracked to abnormal valve development, making it imperative to completely understand the process and regulation of heart valve development. Epithelial-to-mesenchymal transition (EMT) plays an important role in the development of heart valves. Though hemodynamics is vital to valve development, its role in regulating EMT is still unknown. In this study, intracardiac hemodynamics were altered by constricting the outflow tract (OFT)/ventricle junction (OVJ) of HH16–17 (Hamilton and Hamburger (HH) Stage 16–17) chicken embryos, ex ovo for 24 h. The constriction created an increase in peak and time-averaged centerline velocity along the OFT without changes to volumetric flow or heart rate. Computational fluid dynamics was used to estimate the level of increased spatially-averaged wall shear stresses on the OFT cushion from AMIRA reconstructions. OFT constriction led to a significant decrease in OFT cushion volume and the number of invaded mesenchyme in the OFT cushion. qPCR analysis revealed altered mRNA expression of a representative panel of genes, vital to valve development, in the OFT cushions from banded hearts. This study indicates the importance of hemodynamics in valve development.
Collapse
Affiliation(s)
- Vinal Menon
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA; E-Mails: (V.M.); (J.F.E.)
| | - John F. Eberth
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA; E-Mails: (V.M.); (J.F.E.)
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Richard L. Goodwin
- Biomedical Sciences, School of Medicine, University of South Carolina, Greenville, SC 29605, USA; E-Mail:
| | - Jay D. Potts
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA; E-Mails: (V.M.); (J.F.E.)
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-803-216-3820; Fax: +1-803-216-3846
| |
Collapse
|
31
|
Boselli F, Freund JB, Vermot J. Blood flow mechanics in cardiovascular development. Cell Mol Life Sci 2015; 72:2545-59. [PMID: 25801176 PMCID: PMC4457920 DOI: 10.1007/s00018-015-1885-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/25/2015] [Accepted: 03/12/2015] [Indexed: 11/29/2022]
Abstract
Hemodynamic forces are fundamental to development. Indeed, much of cardiovascular morphogenesis reflects a two-way interaction between mechanical forces and the gene network activated in endothelial cells via mechanotransduction feedback loops. As these interactions are becoming better understood in different model organisms, it is possible to identify common mechanogenetic rules, which are strikingly conserved and shared in many tissues and species. Here, we discuss recent findings showing how hemodynamic forces potentially modulate cardiovascular development as well as the underlying fluid and tissue mechanics, with special attention given to the flow characteristics that are unique to the small scales of embryos.
Collapse
Affiliation(s)
- Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France,
| | | | | |
Collapse
|
32
|
Yin L, Wang JP, Xu TP, Chen WM, Huang MD, Xia R, Liu XX, Kong R, Sun M, Zhang EB, Shu YQ. Downregulation of Kruppel-like factor 2 is associated with poor prognosis for nonsmall-cell lung cancer. Tumour Biol 2014; 36:3075-84. [PMID: 25501704 DOI: 10.1007/s13277-014-2943-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/04/2014] [Indexed: 11/27/2022] Open
Abstract
Kruppel-like factor 2 (KLF2) expression is diminished in many malignancies. However, its expression and role in nonsmall-cell lung cancer (NSCLC) remain unknown. In this study, we found that KLF2 levels were decreased in NSCLC tissues compared with adjacent normal tissues. Its expression level was significantly correlated with TNM stages, tumor size, and lymph node metastasis. Moreover, patients with low levels of KLF2 expression had a relatively poor prognosis. Furthermore, knockdown of KLF2 expression by siRNA could promote cell proliferation, while ectopic expression of KLF2 inhibited cell proliferation and promoted apoptosis in NSCLC cells partly via regulating CDKN1A/p21 and CDKN2B/p15 protein expression. Our findings present that decreased KLF2 could be identified as a poor prognostic biomarker in NSCLC and regulate cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Li Yin
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vinjamur DS, Wade KJ, Mohamad SF, Haar JL, Sawyer ST, Lloyd JA. Krüppel-like transcription factors KLF1 and KLF2 have unique and coordinate roles in regulating embryonic erythroid precursor maturation. Haematologica 2014; 99:1565-73. [PMID: 25150253 DOI: 10.3324/haematol.2014.104943] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The Krüppel-like transcription factors KLF1 and KLF2 are essential for embryonic erythropoiesis. They can partially compensate for each other during mouse development, and coordinately regulate numerous erythroid genes, including the β-like globins. Simultaneous ablation of KLF1 and KLF2 results in earlier embryonic lethality and severe anemia. In this study, we determine that this anemia is caused by a paucity of blood cells, and exacerbated by diminished β-like globin gene expression. The anemia phenotype is dose-dependent, and, interestingly, can be ameliorated by a single copy of the KLF2, but not the KLF1 gene. The roles of KLF1 and KLF2 in maintaining normal peripheral blood cell numbers and globin mRNA amounts are erythroid cell-specific. Mechanistic studies led to the discovery that KLF2 has an essential function in erythroid precursor maintenance. KLF1 can partially compensate for KLF2 in this role, but is uniquely crucial for erythroid precursor proliferation through its regulation of G1- to S-phase cell cycle transition. A more drastic impairment of primitive erythroid colony formation from embryonic progenitor cells occurs with simultaneous loss of KLF1 and KLF2 than with loss of a single factor. KLF1 and KLF2 coordinately regulate several proliferation-associated genes, including Foxm1. Differential expression of FoxM1, in particular, correlates with the observed KLF1 and KLF2 gene dosage effects on anemia. Furthermore, KLF1 binds to the FoxM1 gene promoter in blood cells. Thus KLF1 and KLF2 coordinately regulate embryonic erythroid precursor maturation through the regulation of multiple homeostasis-associated genes, and KLF2 has a novel and essential role in this process.
Collapse
Affiliation(s)
- Divya S Vinjamur
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Kristen J Wade
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Safa F Mohamad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jack L Haar
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Stephen T Sawyer
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Joyce A Lloyd
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
34
|
Tsai TN, Lin WS, Wu CH, Lin WY, Chu KM, Cheng CC, Hsu CH, Tsai WC, Cheng SM, Yang SP. Activation of Krüppel-Like Factor 2 with Ginkgo Biloba Extract Induces eNOS Expression and Increases NO Production in Cultured Human Umbilical Endothelial Cells. ACTA CARDIOLOGICA SINICA 2014; 30:215-222. [PMID: 27122791 PMCID: PMC4804860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/24/2013] [Indexed: 06/05/2023]
Abstract
BACKGROUND The mechanisms responsible for the effects of Ginkgo biloba extract (GbE) are not fully understood. Krüppel-like factor 2 (KLF2), a zinc transcription factor, has vasculoprotective effects if activated. The present study attempted to explore whether GbE may activate KLF2 and its consequences. METHODS To determine the effects of GbE on endothelial cells, human umbilical vein endothelial cells (HUVECs) were incubated with various concentrations of GbE. KLF2 expression levels were determined by quantitative reverse transcription polymerase chain reaction. Cytoskeleton staining and cell migration assays were performed to determine the effects of KLF2 activation. Moreover, endothelial NO synthase (eNOS) expression levels were detected by PCR and Western blot testing. Nitric oxide (NO) production was also measured with 4,5-diaminofluorescein. A knockdown of KLF2 was performed to identify the role of KLF2 in GbE-induced eNOS expression and NO production. RESULTS HUVECs that were incubated with GbE increased KLF2 expression. These cells demonstrated an altered cell morphology, cytoskeleton rearrangement, and inhibited migration activity. Moreover, eNOS expression and NO production increased in a dose-dependent manner when cells were treated with GbE. Correspondingly, silencing of KLF2 in HUVECs decreased eNOS expression and NO production in GbE-treated cells. CONCLUSIONS GbE significantly activated KLF2 expression and KLF2-related endothelial function, including cytoskeleton rearrangement, inhibition of migration, eNOS activation, and NO production. These findings suggest that GbE may induce a vasculoprotective effect in endothelial cells. KEY WORDS Endothelial cells; eNOS; Ginkgo biloba extract; KLF2; NO.
Collapse
Affiliation(s)
- Tsung-Neng Tsai
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Shing Lin
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Hsien Wu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Yu Lin
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kai-Min Chu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chung Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hsueng Hsu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Che Tsai
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Meng Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ping Yang
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
35
|
A Pou5f1/Oct4 dependent Klf2a, Klf2b, and Klf17 regulatory sub-network contributes to EVL and ectoderm development during zebrafish embryogenesis. Dev Biol 2014; 385:433-47. [DOI: 10.1016/j.ydbio.2013.10.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
|
36
|
Novodvorsky P, Chico TJ. The Role of the Transcription Factor KLF2 in Vascular Development and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:155-88. [DOI: 10.1016/b978-0-12-386930-2.00007-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Chiplunkar AR, Curtis BC, Eades GL, Kane MS, Fox SJ, Haar JL, Lloyd JA. The Krüppel-like factor 2 and Krüppel-like factor 4 genes interact to maintain endothelial integrity in mouse embryonic vasculogenesis. BMC DEVELOPMENTAL BIOLOGY 2013; 13:40. [PMID: 24261709 PMCID: PMC4222490 DOI: 10.1186/1471-213x-13-40] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/20/2013] [Indexed: 11/30/2022]
Abstract
Background Krüppel-like Factor 2 (KLF2) plays an important role in vessel maturation during embryonic development. In adult mice, KLF2 regulates expression of the tight junction protein occludin, which may allow KLF2 to maintain vascular integrity. Adult tamoxifen-inducible Krüppel-like Factor 4 (KLF4) knockout mice have thickened arterial intima following vascular injury. The role of KLF4, and the possible overlapping functions of KLF2 and KLF4, in the developing vasculature are not well-studied. Results Endothelial breaks are observed in a major vessel, the primary head vein (PHV), in KLF2-/-KLF4-/- embryos at E9.5. KLF2-/-KLF4-/- embryos die by E10.5, which is earlier than either single knockout. Gross hemorrhaging of multiple vessels may be the cause of death. E9.5 KLF2-/-KLF4+/- embryos do not exhibit gross hemorrhaging, but cross-sections display disruptions of the endothelial cell layer of the PHV, and these embryos generally also die by E10.5. Electron micrographs confirm that there are gaps in the PHV endothelial layer in E9.5 KLF2-/-KLF4-/- embryos, and show that the endothelial cells are abnormally bulbous compared to KLF2-/- and wild-type (WT). The amount of endothelial Nitric Oxide Synthase (eNOS) mRNA, which encodes an endothelial regulator, is reduced by 10-fold in E9.5 KLF2-/-KLF4-/- compared to KLF2-/- and WT embryos. VEGFR2, an eNOS inducer, and occludin, a tight junction protein, gene expression are also reduced in E9.5 KLF2-/-KLF4-/- compared to KLF2-/- and WT embryos. Conclusions This study begins to define the roles of KLF2 and KLF4 in the embryonic development of blood vessels. It indicates that the two genes interact to maintain an intact endothelial layer. KLF2 and KLF4 positively regulate the eNOS, VEGFR2 and occludin genes. Down-regulation of these genes in KLF2-/-KLF4-/- embryos may result in the observed loss of vascular integrity.
Collapse
Affiliation(s)
- Aditi R Chiplunkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298-0035, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Chen JL, Lu XJ, Zou KL, Ye K. Krüppel-like factor 2 promotes liver steatosis through upregulation of CD36. J Lipid Res 2013; 55:32-40. [PMID: 23861552 DOI: 10.1194/jlr.m039453] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Krüppel-like factor (KLF) family of transcription factors regulates diverse biological processes that include proliferation, differentiation, apoptosis, development, and responses to external stress. In the present study, we aim to investigate the roles of KLF2 in hepatic steatosis. Our results showed that mRNA and protein levels of KLF2 were significantly elevated in livers from obese mice. Adenoviruses-mediated overexpression of KLF2 induced accumulation of triglycerides in C57BL/6 mice, whereas KLF2 silencing ameliorates hepatosteatosis in ob/ob mice. At the molecular level, our data established CD36 as a novel transcriptional target of KLF2. KLF2 upregulated CD36 expression through a consensus binding site on its proximal promoter region. Additionally, the steatotic effect of KLF2 was dramatically inhibited in CD36-null mice. Therefore, our study reveals a novel link between KLF2-induced hepatic triglyceride accumulation and the expression of CD36.
Collapse
Affiliation(s)
- Jin-Lian Chen
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Annemarieke E. Loot
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| | - Ingrid Fleming
- From the Institute for Vascular Signalling, Centre for Molecular Medicine and German Centre for Cardiovascular Research (DZHK) partner site Rhine-Main, Goethe University, Theodor-Stern-Kai, Frankfurt am Main, Germany
| |
Collapse
|