1
|
Qian S, Long Y, Tan G, Li X, Xiang B, Tao Y, Xie Z, Zhang X. Programmed cell death: molecular mechanisms, biological functions, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e70024. [PMID: 39619229 PMCID: PMC11604731 DOI: 10.1002/mco2.70024] [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: 03/18/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Programmed cell death represents a precisely regulated and active cellular demise, governed by a complex network of specific genes and proteins. The identification of multiple forms of programmed cell death has significantly advanced the understanding of its intricate mechanisms, as demonstrated in recent studies. A thorough grasp of these processes is essential across various biological disciplines and in the study of diseases. Nonetheless, despite notable progress, the exploration of the relationship between programmed cell death and disease, as well as its clinical application, are still in a nascent stage. Therefore, further exploration of programmed cell death and the development of corresponding therapeutic methods and strategies holds substantial potential. Our review provides a detailed examination of the primary mechanisms behind apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. Following this, the discussion delves into biological functions and diseases associated dysregulated programmed cell death. Finally, we highlight existing and potential therapeutic targets and strategies focused on cancers and neurodegenerative diseases. This review aims to summarize the latest insights on programmed cell death from mechanisms to diseases and provides a more reliable approach for clinical transformation.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yao Long
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Guolin Tan
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Xiaoguang Li
- Department of Otolaryngology Head and Neck SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear InstituteShanghai Jiao Tong University School of Medicine, Shanghai Key LabShanghaiChina
| | - Bo Xiang
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Furong LaboratoryCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
2
|
Martin M, Gillett K, Whittick P, Wells SM. New Insights on the Formation of the Mitral Valve Chordae Tendineae in Fetal Life. J Cardiovasc Dev Dis 2024; 11:367. [PMID: 39590210 PMCID: PMC11594762 DOI: 10.3390/jcdd11110367] [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: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
There is an increasing understanding that some mitral valve pathologies have developmental origins. The time course of valvulogenesis varies by animal model; in cattle, the branched chordae tendineae architecture becomes fully developed at full term. The mechanism by which chordae tendineae bifurcate during fetal development remains unknown. The current study presents a detailed description of bovine chordae tendineae formation and bifurcation during fetal development. Analysis of Movat Pentachrome-stained histological sections of the developing mitral valve apparatus was accompanied by micro-CT imaging. TEM imaging of chordae branches and common trunks allowed the measurement of collagen fibril diameter distributions. We observed a proteoglycan-rich "transition zone" at the junction between the fetal mitral valve anterior leaflet and chordae tendineae with "perforations" lined by MMP1/2 and Ki-67 expressing endothelial cells. This region also contained clusters of proliferating endothelial cells within the bulk of the tissue. We hypothesize this zone marks a region where chordae tendineae bifurcate during fetal development. In particular, perforations created by localized MMP activity serve as a site for the initiation of a "split" of a single chordae attachment into two. This is supported by TEM results that suggest a similar population of collagen fibrils runs from the branches into a common trunk. A clear understanding of normal mitral valvulogenesis and its signaling mechanisms will be crucial in developing therapeutics and/or tissue-engineered valve replacements.
Collapse
Affiliation(s)
- Meghan Martin
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Kate Gillett
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Parker Whittick
- Medical Sciences Program, Faculties of Science and Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Sarah Melissa Wells
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Medical Sciences Program, Faculties of Science and Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
3
|
Clift CL, Saunders J, Drake RR, Angel PM. Perspectives on pediatric congenital aortic valve stenosis: Extracellular matrix proteins, post translational modifications, and proteomic strategies. Front Cardiovasc Med 2022; 9:1024049. [PMID: 36439995 PMCID: PMC9685993 DOI: 10.3389/fcvm.2022.1024049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
In heart valve biology, organization of the extracellular matrix structure is directly correlated to valve function. This is especially true in cases of pediatric congenital aortic valve stenosis (pCAVS), in which extracellular matrix (ECM) dysregulation is a hallmark of the disease, eventually leading to left ventricular hypertrophy and heart failure. Therapeutic strategies are limited, especially in pediatric cases in which mechanical and tissue engineered valve replacements may not be a suitable option. By identifying mechanisms of translational and post-translational dysregulation of ECM in CAVS, potential drug targets can be identified, and better bioengineered solutions can be developed. In this review, we summarize current knowledge regarding ECM proteins and their post translational modifications (PTMs) during aortic valve development and disease and contributing factors to ECM dysregulation in CAVS. Additionally, we aim to draw parallels between other fibrotic disease and contributions to ECM post-translational modifications. Finally, we explore the current treatment options in pediatrics and identify how the field of proteomics has advanced in recent years, highlighting novel characterization methods of ECM and PTMs that may be used to identify potential therapeutic strategies relevant to pCAVS.
Collapse
Affiliation(s)
- Cassandra L. Clift
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Janet Saunders
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Richard R. Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Peggi M. Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
- *Correspondence: Peggi M. Angel,
| |
Collapse
|
4
|
Lorda-Diez CI, Solis-Mancilla ME, Sanchez-Fernandez C, Garcia-Porrero JA, Hurle JM, Montero JA. Cell senescence, apoptosis and DNA damage cooperate in the remodeling processes accounting for heart morphogenesis. J Anat 2019; 234:815-829. [PMID: 30875434 PMCID: PMC6539749 DOI: 10.1111/joa.12972] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2019] [Indexed: 01/10/2023] Open
Abstract
During embryonic development, organ morphogenesis requires major tissue rearrangements that are tightly regulated at the genetic level. A large number of studies performed in recent decades assigned a central role to programmed cell death for such morphogenetic tissue rearrangements that often sculpt the shape of embryonic organs. However, accumulating evidence indicates that far from being the only factor responsible for sculpting organ morphology, programmed cell death is accompanied by other tissue remodeling events that ensure the outcome of morphogenesis. In this regard, cell senescence has been recently associated with morphogenetic degenerative embryonic processes as an early tissue remodeling event in development of the limbs, kidney and inner ear. Here, we have explored cell senescence by monitoring β‐galactosidase activity during embryonic heart development where programmed cell death is believed to exert an important morphogenetic function. We report the occurrence of extensive cell senescence foci during heart morphogenesis. These foci overlap spatially and temporally with the areas of programmed cell death that are associated with remodeling of the outflow tract to build the roots of the great arteries and with the septation of cardiac cavities. qPCR analysis allowed us to identify a gene expression profile characteristic of the so‐called senescence secretory associated phenotype in the remodeling outflow tract of the embryonic heart. In addition, we confirmed local upregulation of numerous tumor suppressor genes including p21, p53, p63, p73 and Btg2. Interestingly, the areas of cell senescence were also accompanied by intense lysosomal activation and non‐apoptotic DNA damage revealed by γH2AX immunolabeling. Considering the importance of sustained DNA damage as a triggering factor for cell senescence and apoptosis, we propose the coordinated contribution of DNA damage, senescence and apoptotic cell death to assure tissue remodeling in the developing vertebrate heart.
Collapse
Affiliation(s)
- Carlos I Lorda-Diez
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Michelle E Solis-Mancilla
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Cristina Sanchez-Fernandez
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan A Garcia-Porrero
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan M Hurle
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| | - Juan A Montero
- Facultad de Medicina, Departamento de Anatomía y Biología Celular and IDIVAL, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
5
|
Ma S, Zhao Y, Cao M, Sun C. Human ether‑à‑go‑go‑related gene mutation L539fs/47‑hERG leads to cell apoptosis through the endoplasmic reticulum stress pathway. Int J Mol Med 2019; 43:1253-1262. [PMID: 30628647 PMCID: PMC6365044 DOI: 10.3892/ijmm.2019.4049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 12/31/2018] [Indexed: 11/06/2022] Open
Abstract
Congenital long QT syndrome (LQTS) is a cardiac channelopathy that often results in fatal arrhythmias. LQTS mutations not only lead to abnormal myocardial electrical activities but are associated with heart contraction abnormalities, cardiomyopathy and congenital heart defects. In vivo and in vitro studies have found that LQTS mutations are associated with cardiomyocyte apoptosis, cardiac developmental disorders and even embryonic mortality. Cardiac delayed rectifier potassium channel dysfunction due to the human ether‑à‑go‑go‑related gene (hERG) mutation causes congenital LQTS type 2. The majority of LQTS 2 mutations are characterized by mutant protein accumulation in the endoplasmic reticulum (ER). Unfolded or misfolded protein retention in the ER causes an unfolded protein reaction, which is characteristic of ER stress (ERS). Therefore, the present study hypothesized that LQTS mutations can cause cardiac structural abnormalities via ERS‑mediated cardiomyocyte apoptosis. To test this hypothesis, 293 cells were transiently transfected with an L539fs/47‑hERG plasmid to generate an LQTS 2 model. L539fs/47‑hERG is an LQTS 2 mutation, which consists of a 19‑bp deletion at 1619‑1637 and a point mutation at 1692. Using confocal laser scanning microscopy analysis, it was verified that the L539fs/47‑hERG protein was retained in the ER. Hoechst 33342 apoptosis staining indicated that apoptosis was increased in the L539fs/47‑hERG‑transfected cells, and this be reversed by treatment with 4‑phenyl butyric acid. Western blot analysis revealed increased expression levels of the ERS chaperone glucose regulated protein 78 and pro‑apoptotic ERS‑induced factors, including protein kinase R‑like endoplasmic reticulum kinase, eukaryotic translation‑initiation factor‑2α and C/EBP homologous protein, in the L539fs/47‑hERG‑transfected cells. The B‑cell lymphoma (Bcl‑2)‑associated X protein/Bcl‑2 ratio and caspase‑12 were also increased in the mutated cells. These results demonstrate that L539fs/47‑hERG induces cell apoptosis and the potential molecular mechanism involves the activation of ERS and ERS‑mediated cell apoptosis.
Collapse
Affiliation(s)
- Shuting Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Institute of Cardiovascular Channelopathy, Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi 710061, P.R. China
| | - Yun Zhao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Institute of Cardiovascular Channelopathy, Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi 710061, P.R. China
| | - Miaomiao Cao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Institute of Cardiovascular Channelopathy, Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi 710061, P.R. China
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Institute of Cardiovascular Channelopathy, Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
6
|
Gudipaty SA, Conner CM, Rosenblatt J, Montell DJ. Unconventional Ways to Live and Die: Cell Death and Survival in Development, Homeostasis, and Disease. Annu Rev Cell Dev Biol 2018; 34:311-332. [PMID: 30089222 DOI: 10.1146/annurev-cellbio-100616-060748] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Balancing cell death and survival is essential for normal development and homeostasis and for preventing diseases, especially cancer. Conventional cell death pathways include apoptosis, a form of programmed cell death controlled by a well-defined biochemical pathway, and necrosis, the lysis of acutely injured cells. New types of regulated cell death include necroptosis, pyroptosis, ferroptosis, phagoptosis, and entosis. Autophagy can promote survival or can cause death. Newly described processes of anastasis and resuscitation show that, remarkably, cells can recover from the brink of apoptosis or necroptosis. Important new work shows that epithelia achieve homeostasis by extruding excess cells, which then die by anoikis due to loss of survival signals. This mechanically regulated process both maintains barrier function as cells die and matches rates of proliferation and death. In this review, we describe these unconventional ways in which cells have evolved to die or survive, as well as the contributions that these processes make to homeostasis and cancer.
Collapse
Affiliation(s)
- Swapna A Gudipaty
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christopher M Conner
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| | - Jody Rosenblatt
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| |
Collapse
|
7
|
Lee H, Hong Y, Tran Q, Cho H, Kim M, Kim C, Kwon SH, Park S, Park J, Park J. A new role for the ginsenoside RG3 in antiaging via mitochondria function in ultraviolet-irradiated human dermal fibroblasts. J Ginseng Res 2018; 43:431-441. [PMID: 31308815 PMCID: PMC6606973 DOI: 10.1016/j.jgr.2018.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/09/2018] [Indexed: 11/18/2022] Open
Abstract
Background The efficacy of ginseng, the representative product of Korea, and its chemical effects have been well investigated. The ginsenoside RG3 has been reported to exhibit apoptotic, anticancer, and antidepressant-like effects. Methods In this report, the putative effect of RG3 on several cellular function including cell survival, differentiation, development and aging process were evaluated by monitoring each specific marker. Also, mitochondrial morphology and function were investigated in ultraviolet (UV)-irradiated normal human dermal fibroblast cells. Results RG3 treatment increased the expression of extracellular matrix proteins, growth-associated immediate-early genes, and cell proliferation genes in UV-irradiated normal human dermal fibroblast cells. And, RG3 also resulted in enhanced expression of antioxidant proteins such as nuclear factor erythroid 2–related factor-2 and heme oxygenase-1. In addition, RG3 affects the morphology of UV-induced mitochondria and plays a role in protecting mitochondrial dysfunction. Conclusioin RG3 restores mitochondrial adenosine triphosphate (ATP) and membrane potential via its antioxidant effects in skin cells damaged by UV irradiation, leading to an increase in proteins linked with the extracellular matrix, cell proliferation, and antioxidant activity.
Collapse
Affiliation(s)
- Hyunji Lee
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Youngeun Hong
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Quangdon Tran
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyeonjeong Cho
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Minhee Kim
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chaeyeong Kim
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - So Hee Kwon
- Department of Pharmacy, College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - SungJin Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Corresponding author. Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jisoo Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Corresponding author. Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
8
|
Pang KL, Parnall M, Loughna S. Effect of altered haemodynamics on the developing mitral valve in chick embryonic heart. J Mol Cell Cardiol 2017; 108:114-126. [PMID: 28576718 PMCID: PMC5529288 DOI: 10.1016/j.yjmcc.2017.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/23/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022]
Abstract
Intracardiac haemodynamics is crucial for normal cardiogenesis, with recent evidence showing valvulogenesis is haemodynamically dependent and inextricably linked with shear stress. Although valve anomalies have been associated with genetic mutations, often the cause is unknown. However, altered haemodynamics have been suggested as a pathogenic contributor to bicuspid aortic valve disease. Conversely, how abnormal haemodynamics impacts mitral valve development is still poorly understood. In order to analyse altered blood flow, the outflow tract of the chick heart was constricted using a ligature to increase cardiac pressure overload. Outflow tract-banding was performed at HH21, with harvesting at crucial valve development stages (HH26, HH29 and HH35). Although normal valve morphology was found in HH26 outflow tract banded hearts, smaller and dysmorphic mitral valve primordia were seen upon altered haemodynamics in histological and stereological analysis at HH29 and HH35. A decrease in apoptosis, and aberrant expression of a shear stress responsive gene and extracellular matrix markers in the endocardial cushions were seen in the chick HH29 outflow tract banded hearts. In addition, dysregulation of extracellular matrix (ECM) proteins fibrillin-2, type III collagen and tenascin were further demonstrated in more mature primordial mitral valve leaflets at HH35, with a concomitant decrease of ECM cross-linking enzyme, transglutaminase-2. These data provide compelling evidence that normal haemodynamics are a prerequisite for normal mitral valve morphogenesis, and abnormal blood flow could be a contributing factor in mitral valve defects, with differentiation as a possible underlying mechanism.
Collapse
Affiliation(s)
- Kar Lai Pang
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Matthew Parnall
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Siobhan Loughna
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
9
|
Kupsco A, Schlenk D. Oxidative stress, unfolded protein response, and apoptosis in developmental toxicity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:1-66. [PMID: 26008783 DOI: 10.1016/bs.ircmb.2015.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Physiological development requires precise spatiotemporal regulation of cellular and molecular processes. Disruption of these key events can generate developmental toxicity in the form of teratogenesis or mortality. The mechanism behind many developmental toxicants remains unknown. While recent work has focused on the unfolded protein response (UPR), oxidative stress, and apoptosis in the pathogenesis of disease, few studies have addressed their relationship in developmental toxicity. Redox regulation, UPR, and apoptosis are essential for physiological development and can be disturbed by a variety of endogenous and exogenous toxicants to generate lethality and diverse malformations. This review examines the current knowledge of the role of oxidative stress, UPR, and apoptosis in physiological development as well as in developmental toxicity, focusing on studies and advances in vertebrates model systems.
Collapse
Affiliation(s)
- Allison Kupsco
- Environmental Toxicology Program, University of California, Riverside, CA, USA
| | - Daniel Schlenk
- Environmental Toxicology Program, University of California, Riverside, CA, USA; Environmental Sciences, University of California, Riverside, CA, USA
| |
Collapse
|
10
|
Lauriol J, Jaffré F, Kontaridis MI. The role of the protein tyrosine phosphatase SHP2 in cardiac development and disease. Semin Cell Dev Biol 2014; 37:73-81. [PMID: 25256404 DOI: 10.1016/j.semcdb.2014.09.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 09/09/2014] [Accepted: 09/15/2014] [Indexed: 02/06/2023]
Abstract
Congenital heart disease is the most common human developmental disorder, affecting ∼1:100 newborns, and is the primary cause of birth-defect related deaths worldwide. As a major regulator of receptor tyrosine kinase (RTK), cytokine and G-protein coupled receptor signaling, the non-receptor protein tyrosine phosphatase SHP2 plays a critical role in normal cardiac development and function. Indeed, SHP2 participates in a wide variety of cellular functions, including proliferation, survival, differentiation, migration, and cell-cell communication. Moreover, human activating and inactivating mutations of SHP2 are responsible for two related developmental disorders called Noonan and LEOPARD Syndromes, respectively, which are both characterized, in part, by congenital heart defects. Structural, enzymologic, biochemical, and SHP2 mouse model studies have together greatly enriched our knowledge of SHP2 and, as such, have also uncovered the diverse roles for SHP2 in cardiac development, including its contribution to progenitor cell specification, cardiac morphogenesis, and maturation of cardiac valves and myocardial chambers. By delineating the precise mechanisms by which SHP2 is involved in regulating these processes, we can begin to better understand the pathogenesis of cardiac disease and find more strategic and effective therapies for treatment of patients with congenital heart disorders.
Collapse
Affiliation(s)
- Jessica Lauriol
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Fabrice Jaffré
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Maria I Kontaridis
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
11
|
Abstract
Programmed cell death (PCD) plays a fundamental role in animal development and tissue homeostasis. Abnormal regulation of this process is associated with a wide variety of human diseases, including immunological and developmental disorders, neurodegeneration, and cancer. Here, we provide a brief historical overview of the field and reflect on the regulation, roles, and modes of PCD during animal development. We also discuss the function and regulation of apoptotic proteins, including caspases, the key executioners of apoptosis, and review the nonlethal functions of these proteins in diverse developmental processes, such as cell differentiation and tissue remodeling. Finally, we explore a growing body of work about the connections between apoptosis, stem cells, and cancer, focusing on how apoptotic cells release a variety of signals to communicate with their cellular environment, including factors that promote cell division, tissue regeneration, and wound healing.
Collapse
|
12
|
Currier DG, Polk RC, Reeves RH. A Sonic hedgehog (Shh) response deficit in trisomic cells may be a common denominator for multiple features of Down syndrome. PROGRESS IN BRAIN RESEARCH 2012; 197:223-36. [PMID: 22541295 PMCID: PMC4405118 DOI: 10.1016/b978-0-444-54299-1.00011-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hedgehog (HH) family of growth factors is involved in many aspects of growth and development, from the establishment of left-right axes at gastrulation to the patterning and formation of multiple structures in essentially every tissue, to the maintenance and regulation of stem cell populations in adults. Sonic hedgehog (Shh) in particular acts as a mitogen, regulating proliferation of target cells, a growth factor that triggers differentiation in target populations, and a morphogen causing cells to respond differently based on their positions along a spatial and temporal concentration gradient. Given its very broad range of effects in development, it is not surprising that many of the structures affected by a disruption in Shh signaling are also affected in Down syndrome (DS). However, recent studies have shown that trisomic cerebellar granule cell precursors have a deficit, compared to their euploid counterparts, in their response to the mitogenic effects of Shh. This deficit substantially contributes to the hypocellular cerebellum in mouse models that parallels the human DS phenotype and can be corrected in early development by a single exposure to a small-molecule agonist of the Shh pathway. Here, we consider how an attenuated Shh response might affect several aspects of development to produce multiple phenotypic outcomes observed in DS.
Collapse
Affiliation(s)
- Duane G. Currier
- Department of Physiology and The McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Renita C. Polk
- Department of Physiology and The McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Roger H. Reeves
- Department of Physiology and The McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
13
|
Azhar M, Brown K, Gard C, Chen H, Rajan S, Elliott DA, Stevens MV, Camenisch TD, Conway SJ, Doetschman T. Transforming growth factor Beta2 is required for valve remodeling during heart development. Dev Dyn 2011; 240:2127-41. [PMID: 21780244 DOI: 10.1002/dvdy.22702] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2011] [Indexed: 01/31/2023] Open
Abstract
Although the function of transforming growth factor beta2 (TGFβ2) in epithelial mesenchymal transition (EMT) is well studied, its role in valve remodeling remains to be fully explored. Here, we used histological, morphometric, immunohistochemical and molecular approaches and showed that significant dysregulation of major extracellular matrix (ECM) components contributed to valve remodeling defects in Tgfb2(-/-) embryos. The data indicated that cushion mesenchymal cell differentiation was impaired in Tgfb2(-/-) embryos. Hyaluronan and cartilage link protein-1 (CRTL1) were increased in hyperplastic valves of Tgfb2(-/-) embryos, indicating increased expansion and diversification of cushion mesenchyme into the cartilage cell lineage during heart development. Finally, Western blot and immunohistochemistry analyses indicate that the activation of SMAD2/3 was decreased in Tgfb2(-/-) embryos during valve remodeling. Collectively, the data indicate that TGFβ2 promotes valve remodeling and differentiation by inducing matrix organization and suppressing cushion mesenchyme differentiation into cartilage cell lineage during heart development.
Collapse
Affiliation(s)
- Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, Arizona; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Altunkaynak ME, Altunkaynak BZ, Unal D, Yildirim S, Can I, Unal B. Stereological and histological analysis of the developing rat heart. Anat Histol Embryol 2011; 40:402-10. [PMID: 21569077 DOI: 10.1111/j.1439-0264.2011.01085.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We studied with quantitative and microscopical methods the heart of rats divided into five age groups: embryos at the age of 11 days, fetuses at the age of 16 days and 20 days and also heart samples of 3-day-old pups and young adults (5 weeks of age) were used (n = 10 samples in each group). At the end of the study; heart samples were obtained from all animals. Stereological estimations were performed on heart volume, volume of heart lumen (ventricles and atria), volume of myocardium, numerical density of the myocyte nuclei and mean nuclear diameter of myocytes. Volumetric values and numerical data were estimated via Cavalieri method and physical dissector, respectively. In this study, histological examination was performed at light and electron microscopic levels. The numerical density of the myocyte nuclei increased from fetuses to young adults. Differences between embryos and fetuses, between fetuses and 3-day-old pups, and between 3-day-old pups and young adults were statistically significant. These results indicate that myogenesis continued in the rat myocardium during prenatal life and after birth.
Collapse
Affiliation(s)
- M E Altunkaynak
- Department of Histology and Embryology, Medical School, Ondokuz Mayıs University, Samsun, Turkey
| | | | | | | | | | | |
Collapse
|
15
|
Song L, Zhao M, Wu B, Zhou B, Wang Q, Jiao K. Cell autonomous requirement of endocardial Smad4 during atrioventricular cushion development in mouse embryos. Dev Dyn 2011; 240:211-20. [PMID: 21089072 DOI: 10.1002/dvdy.22493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrioventricular (AV) cushions are the precursors of AV septum and valves. In this study, we examined roles of Smad4 during AV cushion development using a conditional gene inactivation approach. We found that endothelial/endocardial inactivation of Smad4 led to the hypocellular AV cushion defect and that both reduced cell proliferation and increased apoptosis contributed to the defect. Expression of multiple genes critical for cushion development was down-regulated in mutant embryos. In collagen gel assays, the number of mesenchymal cells formed is significantly reduced in mutant AV explants compared to that in control explants, suggesting that the reduction of cushion mesenchyme formation in mutants is unlikely secondary to their gross vasculature abnormalities. Using a previously developed immortal endocardial cell line, we showed that Smad4 is required for BMP signaling- stimulated upregulation of Tbx20 and Gata4. Therefore, our data collectively support the cell-autonomous requirement of endocardial Smad4 in regulating AV cushion development.
Collapse
Affiliation(s)
- Langying Song
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
16
|
Jiang HK, Qiu GR, Li-Ling J, Xin N, Sun KL. Reduced ACTC1 expression might play a role in the onset of congenital heart disease by inducing cardiomyocyte apoptosis. Circ J 2010; 74:2410-8. [PMID: 20962418 DOI: 10.1253/circj.cj-10-0234] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The Cardiac α actin 1 gene (ACTC1) has been related to familial atrial septal defects. This study was set to explore a potential role of this gene in the formation of sporadic congenital heart disease (CHD). METHODS AND RESULTS Assessment of cardiac tissue samples from 33 patients with sporadic CHD (gestational age (GA) 18 weeks-49 months) with real-time RT-PCR, Western blotting and immunohistochemistry has revealed a markedly decreased ACTC1 expression in the majority of samples (78.8%) compared with autopsied normal heart tissue from aged-matched subjects (GA 17 weeks-36 months). Also, as shown by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assay, the proportion of apoptotic cardiomyocytes in samples featuring down-regulated ACTC1 expression (Group 1) was significantly greater than those with normal expression (Group 2) and the controls (P<0.01). The proportion of apoptotic cells strongly correlated with the expression of ACTC1 (r=-0.918, P<0.01). A study of 2 essential genes involved in apoptosis, Caspase-3 and Bcl-2, confirmed that the former has significantly increased expression, whilst the latter has decreased expression in Group 1 than in the other groups (P<0.01). Transfection of a small interfering RNA targeting, Actc1 (Actc1-siRNA), to a cardiomyocyte cell line, H9C2, also detected more apoptotic cells. CONCLUSIONS Reduced ACTC1 expression might play a role in the onset of CHD through induction of cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Hong-Kun Jiang
- Department of Pediatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
17
|
Azhar M, Runyan RB, Gard C, Sanford LP, Miller ML, Andringa A, Pawlowski S, Rajan S, Doetschman T. Ligand-specific function of transforming growth factor beta in epithelial-mesenchymal transition in heart development. Dev Dyn 2009; 238:431-42. [PMID: 19161227 DOI: 10.1002/dvdy.21854] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The ligand specificity of transforming growth factor beta (TGFbeta) in vivo in mouse cardiac cushion epithelial-to-mesenchymal transition (EMT) is poorly understood. To elucidate the function of TGFbeta in cushion EMT, we analyzed Tgfb1(-/-), Tgfb2(-/-), and Tgfb3(-/-) mice between embryonic day (E) 9.5 and E14.5 using both in vitro and in vivo approaches. Atrioventricular (AV) canal collagen gel assays at E9.5 indicated normal EMT in both Tgfb1(-/-) and Tgfb3(-/-) mice. However, analysis of Tgfb2(-/-) AV explants at E9.5 and E10.5 indicated that EMT, but not cushion cell proliferation, was initially delayed but later remained persistent. This was concordant with the observation that Tgfb2(-/-) embryos, and not Tgfb1(-/-) or Tgfb3(-/-) embryos, develop enlarged cushions at E14.5 with elevated levels of well-validated indicators of EMT. Collectively, these data indicate that TGFbeta2, and not TGFbeta1 or TGFbeta3, mediates cardiac cushion EMT by promoting both the initiation and cessation of EMT.
Collapse
Affiliation(s)
- Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Barth M, Schumacher H, Kuhn C, Akhyari P, Lichtenberg A, Franke WW. Cordial connections: molecular ensembles and structures of adhering junctions connecting interstitial cells of cardiac valves in situ and in cell culture. Cell Tissue Res 2009; 337:63-77. [PMID: 19475424 DOI: 10.1007/s00441-009-0806-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 04/06/2009] [Indexed: 01/19/2023]
Abstract
Remarkable efforts have recently been made in the tissue engineering of heart valves to improve the results of valve transplantations and replacements, including the design of artificial valves. However, knowledge of the cell and molecular biology of valves and, specifically, of valvular interstitial cells (VICs) remains limited. Therefore, our aim has been to determine and localize the molecules forming the adhering junctions (AJs) that connect VICs in situ and in cell culture. Using biochemical and immunolocalization methods at the light- and electron-microscopic levels, we have identified, in man, cow, sheep and rat, the components of VIC-connecting AJs in situ and in cell culture. These AJs contain, in addition to the transmembrane glycoproteins N-cadherin and cadherin-11, the typical plaque proteins alpha- and beta-catenin as well as plakoglobin and p120, together with minor amounts of protein p0071, i.e. a total of five plaque proteins of the armadillo family. While we can exclude the occurrence of desmogleins, desmocollins and desmoplakin, we have noted with surprise that AJs of VICs in cell cultures, but not those growing in the valve tissue, contain substantial amounts of the desmosomal plaque protein, plakophilin-2. Clusters of AJs occur not only on the main VIC cell bodies but are also found widely dispersed on their long filopodia thus forming, in the tissue, a meshwork that, together with filopodial attachments to paracrystalline collagen fiber bundles, establishes a three-dimensional suprastructure, the role of which is discussed with respect to valve formation, regeneration and function.
Collapse
Affiliation(s)
- Mareike Barth
- Helmholtz Group/Cell Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Snarr BS, Wirrig EE, Phelps AL, Trusk TC, Wessels A. A spatiotemporal evaluation of the contribution of the dorsal mesenchymal protrusion to cardiac development. Dev Dyn 2007; 236:1287-94. [PMID: 17265457 PMCID: PMC4003411 DOI: 10.1002/dvdy.21074] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The mesenchymal tissues involved in cardiac septation are derived from different sources. In addition to endocardial-derived mesenchyme, the heart also receives contributions from the neural crest, the proepicardium, and the dorsal mesenchymal protrusion (DMP). Whereas the contributions of the neural crest and proepicardium have been thoroughly studied, the DMP has received little attention. Here, we present the results of a comprehensive spatiotemporal study of the DMP in cardiac development. Using the Tie2-Cre mouse, immunohistochemistry, and AMIRA reconstructions, we show that the DMP, in combination with the mesenchymal cap on the primary atrial septum, fuse with the major atrioventricular cushions to close the primary atrial foramen and to form the atrioventricular mesenchymal complex. In this complex, the DMP constitutes a discrete prominent mesenchymal component, wedged in between the major cushions. This new model for atrioventricular septation may provide novel insights into understanding the etiology of congenital cardiac malformations.
Collapse
Affiliation(s)
- Brian S. Snarr
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina
| | - Elaine E. Wirrig
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina
| | - Aimee L. Phelps
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina
| | - Thomas C. Trusk
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina
| | - Andy Wessels
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, Charleston, South Carolina
- Correspondence to: Andy Wessels, Ph.D., Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building, Room 648B, P.O. Box 250508, Charleston, SC 29425.
| |
Collapse
|
20
|
Zwerts F, Lupu F, De Vriese A, Pollefeyt S, Moons L, Altura RA, Jiang Y, Maxwell PH, Hill P, Oh H, Rieker C, Collen D, Conway SJ, Conway EM. Lack of endothelial cell survivin causes embryonic defects in angiogenesis, cardiogenesis, and neural tube closure. Blood 2007; 109:4742-52. [PMID: 17299096 PMCID: PMC1885533 DOI: 10.1182/blood-2006-06-028068] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 02/07/2007] [Indexed: 11/20/2022] Open
Abstract
We explored the physiologic role of endothelial cell apoptosis during development by generating mouse embryos lacking the inhibitor of apoptosis protein (IAP) survivin in endothelium. This was accomplished by intercrossing survivin(lox/lox) mice with mice expressing cre recombinase under the control of the endothelial cell specific tie1 promoter (tie1-cre mice). Lack of endothelial cell survivin resulted in embryonic lethality. Mutant embryos had prominent and diffuse hemorrhages from embryonic day 9.5 (E9.5) and died before E13.5. Heart development was strikingly abnormal. Survivin-null endocardial lineage cells could not support normal epithelial-mesenchymal transformation (EMT), resulting in hypoplastic endocardial cushions and in utero heart failure. In addition, 30% of mutant embryos had neural tube closure defects (NTDs) that were not caused by bleeding or growth retardation, but were likely due to alterations in the release of soluble factors from endothelial cells that otherwise support neural stem cell proliferation and neurulation. Thus, regulation of endothelial cell survival, and maintenance of vascular integrity by survivin are crucial for normal embryonic angiogenesis, cardiogenesis, and neurogenesis.
Collapse
Affiliation(s)
- Femke Zwerts
- Center for Transgene Technology and Gene Therapy, University of Leuven, Gasthuisberg O&N1, Herestraat 49, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Stephen LJ, Fawkes AL, Verhoeve A, Lemke G, Brown A. A critical role for the EphA3 receptor tyrosine kinase in heart development. Dev Biol 2007; 302:66-79. [PMID: 17046737 DOI: 10.1016/j.ydbio.2006.08.058] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/23/2006] [Accepted: 08/24/2006] [Indexed: 10/24/2022]
Abstract
Eph proteins are receptor tyrosine kinases that control changes in cell shape and migration during development. We now describe a critical role for EphA3 receptor signaling in heart development as revealed by the phenotype of EphA3 null mice. During heart development mesenchymal outgrowths, the atrioventricular endocardial cushions, form in the atrioventricular canal. This morphogenetic event requires endocardial cushion cells to undergo an epithelial to mesenchymal transformation (EMT), and results in the formation of the atrioventricular valves and membranous portions of the atrial and ventricular septa. We show that EphA3 knockouts have significant defects in the development of their atrial septa and atrioventricular endocardial cushions, and that these cardiac abnormalities lead to the death of approximately 75% of homozygous EphA3(-/-) mutants. We demonstrate that EphA3 and its ligand, ephrin-A1, are expressed in adjacent cells in the developing endocardial cushions. We further demonstrate that EphA3(-/-) atrioventricular endocardial cushions are hypoplastic compared to wildtype and that EphA3(-/-) endocardial cushion explants give rise to fewer migrating mesenchymal cells than wildtype explants. Thus our results indicate that EphA3 plays a crucial role in the development and morphogenesis of the cells that give rise to the atrioventricular valves and septa.
Collapse
Affiliation(s)
- Lesley J Stephen
- BioTherapeutics Research Group, The John P Robarts Research Institute, and The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
22
|
Song L, Fässler R, Mishina Y, Jiao K, Baldwin HS. Essential functions of Alk3 during AV cushion morphogenesis in mouse embryonic hearts. Dev Biol 2007; 301:276-86. [PMID: 16959237 DOI: 10.1016/j.ydbio.2006.08.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 07/11/2006] [Accepted: 08/01/2006] [Indexed: 11/22/2022]
Abstract
Accumulated evidence has suggested that BMP pathways play critical roles during mammalian cardiogenesis and impairment of BMP signaling may contribute to human congenital heart diseases (CHDs), which are the leading cause of infant morbidity and mortality. Alk3 encodes a BMP specific type I receptor expressed in mouse embryonic hearts. To reveal functions of Alk3 during atrioventricular (AV) cushion morphogenesis and to overcome the early lethality of Alk3(-/-) embryos, we applied a Cre/loxp approach to specifically inactivate Alk3 in the endothelium/endocardium. Our studies showed that endocardial depletion of Alk3 severely impairs epithelium-mesenchymal-transformation (EMT) in the atrioventricular canal (AVC) region; the number of mesenchymal cells formed in Tie1-Cre;Alk3(loxp/loxp) embryos was reduced to only approximately 20% of the normal level from both in vivo section studies and in vitro explant assays. We showed, for the first time, that in addition to its functions on mesenchyme formation, Alk3 is also required for the normal growth/survival of AV cushion mesenchymal cells. Functions of Alk3 are accomplished through regulating expression/activation/subcellular localization of multiple downstream genes including Smads and cell-cycle regulators. Taken together, our study supports the notion that Alk3-mediated BMP signaling in AV endocardial/mesenchymal cells plays a central role during cushion morphogenesis.
Collapse
Affiliation(s)
- Lanying Song
- Division of Genetic and Translational Medicine, Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
23
|
Flagg AE, Earley JU, Svensson EC. FOG-2 attenuates endothelial-to-mesenchymal transformation in the endocardial cushions of the developing heart. Dev Biol 2006; 304:308-16. [PMID: 17274974 PMCID: PMC1868509 DOI: 10.1016/j.ydbio.2006.12.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2006] [Revised: 12/04/2006] [Accepted: 12/17/2006] [Indexed: 11/30/2022]
Abstract
Development of the heart valves is a complex process that relies on the successful remodeling of endocardial cushions. This process is dependent on a number of transcriptional regulators, including GATA4 and its interacting partner FOG-2. We have previously shown that the endocardial cushions in FOG-2 deficient mice are hyperplastic and fail to remodel appropriately, suggesting a defect late in endocardial cushion development. To elucidate this defect, we examined the later steps in endocardial cushion development including mesenchymal cell proliferation, differentiation, and apoptosis. We also measured myocardialization and endothelial-to-mesenchymal transformation (EMT) using previously described in vitro assays. We found no difference in the ability of the endocardial cushions to undergo myocardialization or in the rates of mesenchymal cell proliferation, differentiation, or apoptosis in the FOG-2 deficient cushions when compared to wild-type controls. However, using a collagen gel invasion assay, we found a 78% increase in outflow tract cushion EMT and a 35% increase in atrioventricular cushion EMT in the FOG-2 deficient mice when compared with wild-type mice. Taken together with GATA4's known role in promoting EMT, these results suggest that FOG-2 functions in cardiac valve formation as an attenuator of EMT by repressing GATA4 activity within the developing endocardial cushions.
Collapse
Affiliation(s)
- Alleda E Flagg
- Department of Medicine, University of Chicago, Section of Cardiology, 5841 S. Maryland Ave, MC6088, Chicago, IL 60637, USA
| | | | | |
Collapse
|
24
|
Abstract
The matricellular protein CCN1 (formerly named CYR61) regulates cell adhesion, migration, proliferation, survival, and differentiation through binding to integrin receptors and heparan sulfate proteoglycans. Here we show that Ccn1-null mice are impaired in cardiac valvuloseptal morphogenesis, resulting in severe atrioventricular septal defects (AVSD). Remarkably, haploinsufficiency for Ccn1 also results in delayed formation of the ventricular septum in the embryo and persistent ostium primum atrial septal defects (ASD) in approximately 20% of adults. Mechanistically, Ccn1 is not required for epithelial-to-mesenchymal transformation or cell proliferation and differentiation in the endocardial cushion tissue. However, Ccn1 deficiency leads to precocious apoptosis in the atrial junction of the cushion tissue and impaired gelatinase activities in the muscular component of the interventricular septum at embryonic day 12.5, when fusion between the endocardial cushion tissue and the atrial and ventricular septa occurs, indicating that these defects may underlie the observed AVSD. Moreover, human CCN1 maps to 1p21-p31, the chromosomal location of an AVSD susceptibility gene. Together, these results provide evidence that deficiency in matrix signaling can lead to autosomal dominant AVSD, identify Ccn1(+/-) mice as a genetic model for ostium primum ASD, and implicate CCN1 as a candidate gene for AVSD in humans.
Collapse
Affiliation(s)
| | - Lester F. Lau
- *Corresponding Author: Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, IL 60607. Phone: (312) 996-6978; Fax: (312) 996-7034;
| |
Collapse
|
25
|
Penaloza C, Lin L, Lockshin RA, Zakeri Z. Cell death in development: shaping the embryo. Histochem Cell Biol 2006; 126:149-58. [PMID: 16816938 DOI: 10.1007/s00418-006-0214-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2006] [Indexed: 01/13/2023]
Abstract
Cell death in animals is normally classified as type I (apoptotic), type II (autophagic) or necrotic. Of the biologically controlled types of death, in most embryos apoptosis is the most common, although in metamorphosis and in cells with massive cytoplasm type II is often seen, and intermediate forms are seen. For vertebrate embryos other than mammals, apoptosis is not seen prior to gastrulation but thereafter is used to sculpt the organs of the embryo, while overproduction of cells with subsequent death of excess cells is a common means of generating high specificity with low information cost. In zebrafish at least, the inability of embryos prior to the maternal-zygotic transition to undergo apoptosis appears to derive from the inability of the cells to resist lysis once apoptosis begins, rather than any inhibition of apoptosis. In mammalian embryos, apoptosis is seen during cavitation. Thereafter, as in other embryos, cell death plays a major role in shaping and sculpting the embryo. In those situations that have been carefully studied, cell death is under tight genetic control (including regulation of gene products whose function in cell death is not yet known, such as cdk5), with activation of apoptosis sometimes regulated by local environmental variables.
Collapse
Affiliation(s)
- Carlos Penaloza
- Department of Biology, Queens College and Graduate Center of CUNY, Flushing, NY 11367, USA
| | | | | | | |
Collapse
|
26
|
Hinton RB, Lincoln J, Deutsch GH, Osinska H, Manning PB, Benson DW, Yutzey KE. Extracellular Matrix Remodeling and Organization in Developing and Diseased Aortic Valves. Circ Res 2006; 98:1431-8. [PMID: 16645142 DOI: 10.1161/01.res.0000224114.65109.4e] [Citation(s) in RCA: 324] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Heart valve disease is an important cause of morbidity and mortality worldwide. Little is known about valve disease pathogenesis, but increasing evidence implicates a genetic basis for valve disease, suggesting a developmental origin. Although the cellular and molecular processes involved in early valvulogenesis have been well described, less is known about the regulation of valve extracellular matrix (ECM) organization and valvular interstitial cell (VIC) distribution that characterize the mature valve structure. Histochemistry, immunohistochemistry, and electron microscopy were used to examine ECM organization, VIC distribution, and cell proliferation during late valvulogenesis in chicken and mouse. In mature valves, ECM organization is conserved across species, and developmental studies demonstrate that ECM stratification begins during late embryonic cusp remodeling and continues into postnatal life. Cell proliferation decreases concomitant with ECM stratification and VIC compartmentalization. Explanted, stenotic bicuspid aortic valves (BAVs) from pediatric patients were also examined. The diseased valves exhibited disruption of the highly organized ECM and VIC distribution seen in normal valves. Cusps from diseased valves were thickened with increased and disorganized collagens and proteoglycans, decreased and fragmented elastic fibers, and cellular disarray without calcification or cell proliferation. Taken together, these studies show that normal valve development is characterized by spatiotemporal coordination of ECM organization and VIC compartmentalization and that these developmental processes are disrupted in pediatric patients with diseased BAVs.
Collapse
Affiliation(s)
- Robert B Hinton
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang H, Zhou L, Yang R, Sheng Y, Sun W, Kong X, Cao K. Identification of differentially expressed genes in human heart with ventricular septal defect using suppression subtractive hybridization. Biochem Biophys Res Commun 2006; 342:135-44. [PMID: 16472770 DOI: 10.1016/j.bbrc.2006.01.113] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2006] [Accepted: 01/23/2006] [Indexed: 11/29/2022]
Abstract
Ventricular septal defect (VSD) accounts for the largest number of birth congenital heart defects in human, but the genetic programs that control ventricular septation are poorly understood. To identify differentially expressed genes between ventricular septal defect and normal ventricular septum myocardium, we have undertaken suppression subtractive hybridization (SSH) and generated reciprocal cDNA collections of representative mRNAs specific to human heart with ventricular septal defect versus normal control. Following SSH, 1378 clones were sequenced and found to derive from 551 different genes. These predominately expressed genes included genes involved in energy metabolism, cell cycle and growth, cytoskeleton and cell adhesion, LIM protein, zinc finger protein, and development. It is anticipated that further study of genes identified will provide insights into their specific roles in the etiology of VSD, even in cardiac development, aging, and disease.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, PR China
| | | | | | | | | | | | | |
Collapse
|
28
|
Kokubo H, Miyagawa-Tomita S, Johnson RL. Hesr, a Mediator of the Notch Signaling, Functions in Heart and Vessel Development. Trends Cardiovasc Med 2005; 15:190-4. [PMID: 16165016 DOI: 10.1016/j.tcm.2005.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Revised: 05/12/2005] [Accepted: 05/13/2005] [Indexed: 11/19/2022]
Abstract
Hesr genes are members of the hairy and enhancer of split-related (hesr) gene family of basic helix-loop-helix-type transcriptional repressors. hesr genes have been implicated in cardiovascular development as the primary targets of Notch signaling. Functional analysis of hesr2 knockout mice revealed abnormal cardiac hemodynamics, such as atrioventricular valve regurgitation and reduced left ventricular systolic function, caused by hypoplastic AV valves and abnormal cardiomyocytes. Recent evidence demonstrates that hesr1 and hesr2 function redundantly in epithelial-to-mesenchymal transformation during atrioventricular valve formation and maintenance of trabecular cells in the heart ventricles, and in arterial-venous differentiation of blood vessels. This review highlights the many functions of the hesr gene family in heart and vessel development.
Collapse
Affiliation(s)
- Hiroki Kokubo
- Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan.
| | | | | |
Collapse
|
29
|
Person AD, Garriock RJ, Krieg PA, Runyan RB, Klewer SE. Frzb modulates Wnt-9a-mediated beta-catenin signaling during avian atrioventricular cardiac cushion development. Dev Biol 2005; 278:35-48. [PMID: 15649459 DOI: 10.1016/j.ydbio.2004.10.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 10/08/2004] [Accepted: 10/21/2004] [Indexed: 10/26/2022]
Abstract
Normal development of the cardiac atrioventricular (AV) endocardial cushions is essential for proper ventricular septation and morphogenesis of the mature mitral and tricuspid valves. In this study, we demonstrate spatially restricted expression of both Wnt-9a (formerly Wnt-14) and the secreted Wnt antagonist Frzb in AV endocardial cushions of the developing chicken heart. Wnt-9a expression is detected only in AV canal endocardial cells, while Frzb expression is detected in both endocardial and transformed mesenchymal cells of the developing AV cardiac cushions. We present evidence that Wnt-9a promotes cell proliferation in the AV canal and overexpression of Wnt-9a in ovo results in enlarged endocardial cushions and AV inlet obstruction. Wnt-9a stimulates beta-catenin-responsive transcription in AV canal cells, duplicates the embryonic axis upon ventral injections in Xenopus embryos and appears to regulate cell proliferation by activating a Wnt/beta-catenin signaling pathway. Additional functional studies reveal that Frzb inhibits Wnt-9a-mediated cell proliferation in cardiac cushions. Together, these data argue that Wnt-9a and Frzb regulate mesenchymal cell proliferation leading to proper AV canal cushion outgrowth and remodeling in the developing avian heart.
Collapse
Affiliation(s)
- Anthony D Person
- Department of Cell Biology and Anatomy, University Medical Center, University of Arizona School of Medicine, 1501 N. Campbell Avenue, PO Box 245044, Tucson, AZ 85724, USA.
| | | | | | | | | |
Collapse
|
30
|
Hallaq H, Pinter E, Enciso J, McGrath J, Zeiss C, Brueckner M, Madri J, Jacobs HC, Wilson CM, Vasavada H, Jiang X, Bogue CW. A null mutation of Hhex results in abnormal cardiac development, defective vasculogenesis and elevated Vegfa levels. Development 2004; 131:5197-209. [PMID: 15459110 DOI: 10.1242/dev.01393] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The homeobox gene Hhex has recently been shown to be essential for normal liver, thyroid and forebrain development. Hhex(-/-) mice die by mid-gestation (E14.5) and the cause of their early demise remains unclear. Because Hhex is expressed in the developing blood islands at E7.0 in the endothelium of the developing vasculature and heart at E9.0-9.5, and in the ventral foregut endoderm at E8.5-9.0, it has been postulated to play a critical role in heart and vascular development. We show here, for the first time, that a null mutation of Hhex results in striking abnormalities of cardiac and vascular development which include: (1) defective vasculogenesis, (2) hypoplasia of the right ventricle, (3) overabundant endocardial cushions accompanied by ventricular septal defects, outflow tract abnormalities and atrio-ventricular (AV) valve dysplasia and (4) aberrant development of the compact myocardium. The dramatic enlargement of the endocardial cushions in the absence of Hhex is due to decreased apoptosis and dysregulated epithelial-mesenchymal transformation (EMT). Interestingly, vascular endothelial growth factor A (Vegfa) levels in the hearts of Hhex(-/-) mice were elevated as much as three-fold between E9.5 and E11.5, and treatment of cultured Hhex(-/-) AV explants with truncated soluble Vegfa receptor 1, sFlt-1, an inhibitor of Vegf signaling, completely abolished the excessive epithelial-mesenchymal transformation seen in the absence of Hhex. Therefore, Hhex expression in the ventral foregut endoderm and/or the endothelium is necessary for normal cardiovascular development in vivo, and one function of Hhex is to repress Vegfa levels during development.
Collapse
Affiliation(s)
- Haifa Hallaq
- Department of Pediatrics, Yale University School of Medicine, 464 Congress Avenue, New Haven, CT, 06519-1361, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abdelwahid E, Pelliniemi LJ, Szucsik JC, Lessard JL, Jokinen E. Cellular disorganization and extensive apoptosis in the developing heart of mice that lack cardiac muscle alpha-actin: apparent cause of perinatal death. Pediatr Res 2004; 55:197-204. [PMID: 14605248 DOI: 10.1203/01.pdr.0000100900.56627.e1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mice that lack cardiac muscle alpha-actin die during the perinatal period. Approximately 56% of mice that are homozygous null (-/-) for a functional cardiac alpha-actin gene do not survive to term, and the remainder generally die within 2 wk of birth. We found that there were neither morphologic differences nor differences in the extent of apoptosis between the mutant and normal hearts on embryonic day (E) 12 and E14 of development. However, apoptosis was greater in the hearts of homozygous null mice on E17 and postnatal day 1 when compared with wild-type hearts. The antiapoptotic factor Bcl-x/(L) was localized in regions adjacent to where apoptosis was detected. The distribution patterns of the apoptosis triggering protein p53 were similar to those of apoptotic cells. The growth of the prenatal and postnatal hearts of the cardiac alpha-actin-deficient mice was retarded, and the cytoplasmic filaments were disorganized. Although apoptotic cells were observed in both the atria and ventricles in the hearts of the homozygous null animals, the frequency was greater in the ventricles than in the atria. Our results indicate that the functional and structural disturbances in the mice with a homozygous lack of cardiac alpha-actin seem to be due to disorganized development of acto-myosin filaments in the affected cardiomyocytes. Other actin isoforms cannot compensate for the lack of cardiac alpha-actin, and this seems to induce apoptosis in defective cardiac myocytes, which are not able to cope with the increased workload in the perinatal phase.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- Department of Pediatrics and Medicity Research Laboratories, University of Turku, Finland.
| | | | | | | | | |
Collapse
|
32
|
Gitler AD, Lu MM, Jiang YQ, Epstein JA, Gruber PJ. Molecular markers of cardiac endocardial cushion development. Dev Dyn 2003; 228:643-50. [PMID: 14648841 DOI: 10.1002/dvdy.10418] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Endocardial cushions are precursors of mature heart valves. They form within the looped heart tube as discrete swellings and develop into thin, pliable leaflets that prevent regurgitation of blood. The embryonic origins of cardiac valves include endothelial, myocardial, and neural crest cells. Recently, an increasing number of animal models derived from mutational screens, gene inactivation, and transgenic studies have identified specific molecules required for normal development of the cardiac valves, and critical molecular pathways are beginning to emerge. To further this process, we have sought to assemble a diverse set of molecular markers encompassing all stages of cardiac valve development. Here, we provide a detailed comparative gene expression analysis of thirteen endocardial cushion markers. We identify endocardial cushion expression of the transcription factor Fog1, and we demonstrate active Wnt/beta-catenin signaling in developing endocardial cushions suggesting pathways that have not been previously appreciated to participate in cardiac valve formation.
Collapse
Affiliation(s)
- Aaron D Gitler
- Department of Medicine, Cardiology Division, University of Pennsylvania Health System, 954 BRB II/III, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|