1
|
Wang H, Zhao B, Huang L, Zhu X, Li N, Huang C, Han Z, Ouyang K. Conditional deletion of IP 3R1 by Islet1-Cre in mice reveals a critical role of IP 3R1 in interstitial cells of Cajal in regulating GI motility. J Gastroenterol 2025; 60:152-165. [PMID: 39476178 DOI: 10.1007/s00535-024-02164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/16/2024] [Indexed: 02/05/2025]
Abstract
BACKGROUND AND AIMS Inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) has been proposed to play a physiological role in regulating gastrointestinal (GI) motility, but the underlying cell-dependent mechanism remains unclear. Here, we utilized cell-specific IP3R1 deletion strategies to address this question in mice. METHODS Conditional IP3R1 knockout mice using Wnt1-Cre, Islet1-Cre mice, and smMHC-CreEGFP were generated. Cell lineage tracing was performed to determine where gene deletion occurred in the GI tract. Whole-gut transit assay and isometric tension recording were used to assess GI function in vivo and in vitro. RESULTS In the mouse GI tract, Islet1-Cre targeted smooth muscle cells (SMCs) and interstitial cells of Cajal (ICCs), but not enteric neurons. IP3R1 deletion by Islet1-Cre (isR1KO) caused a phenotype of intestinal pseudo-obstruction (IPO), evidenced by prolonged whole-gut transit time, enlarged GI tract, abdominal distention, and early lethality. IP3R1 deletion by Islet1-Cre not only reduced the frequency of spontaneous contractions but also decreased the contractile responses to the muscarinic agonist carbachol (CCh) and electrical field stimulation (EFS) in colonic circular muscles. By contrast, smMHC-CreEGFP only targeted SMCs in the mouse GI tract. Although IP3R1 deletion by smMHC-CreEGFP (smR1KO) also reduced the contractile responses to CCh and EFS in colonic circular muscles, the frequency of spontaneous contractions was less affected, and neither global GI abnormalities nor early lethality was found in smR1KO mice. CONCLUSIONS IP3R1 deletion in both ICCs and SMCs but not in SMCs alone causes an IPO phenotype, suggesting that IP3R1 in ICCs plays an essential role in regulating GI motility in vivo.
Collapse
Affiliation(s)
- Hong Wang
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China
| | - Beili Zhao
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| | - Kunfu Ouyang
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China.
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| |
Collapse
|
2
|
Piamsiri C, Fefelova N, Pamarthi SH, Gwathmey JK, Chattipakorn SC, Chattipakorn N, Xie LH. Potential Roles of IP 3 Receptors and Calcium in Programmed Cell Death and Implications in Cardiovascular Diseases. Biomolecules 2024; 14:1334. [PMID: 39456267 PMCID: PMC11506173 DOI: 10.3390/biom14101334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle communications during both physiological and pathological processes. Accumulating evidence from in vitro, in vivo, and clinical studies supports the notion that the overactivation of IP3Rs is linked to the pathogenesis of various cardiac conditions. The overactivation of IP3Rs results in the dysregulation of Ca2+ concentration ([Ca2+]) within cytosolic, mitochondrial, and nucleoplasmic cellular compartments. In cardiovascular pathologies, two isoforms of IP3Rs, i.e., IP3R1 and IP3R2, have been identified. Notably, IP3R1 plays a pivotal role in cardiac ischemia and diabetes-induced arrhythmias, while IP3R2 is implicated in sepsis-induced cardiomyopathy and cardiac hypertrophy. Furthermore, IP3Rs have been reported to be involved in various programmed cell death (PCD) pathways, such as apoptosis, pyroptosis, and ferroptosis underscoring their multifaceted roles in cardiac pathophysiology. Based on these findings, it is evident that exploring potential therapeutic avenues becomes crucial. Both genetic ablation and pharmacological intervention using IP3R antagonists have emerged as promising strategies against IP3R-related pathologies suggesting their potential therapeutic potency. This review summarizes the roles of IP3Rs in cardiac physiology and pathology and establishes a foundational understanding with a particular focus on their involvement in the various PCD pathways within the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chanon Piamsiri
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Sri Harika Pamarthi
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| |
Collapse
|
3
|
Huang F, Zhang F, Huang L, Zhu X, Huang C, Li N, Da Q, Huang Y, Yang H, Wang H, Zhao L, Lin Q, Chen Z, Xu J, Liu J, Ren M, Wang Y, Han Z, Ouyang K. Inositol 1,4,5-Trisphosphate Receptors Regulate Vascular Smooth Muscle Cell Proliferation and Neointima Formation in Mice. J Am Heart Assoc 2024; 13:e034203. [PMID: 39023067 PMCID: PMC11964046 DOI: 10.1161/jaha.124.034203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) proliferation is involved in many types of arterial diseases, including neointima hyperplasia, in which Ca2+ has been recognized as a key player. However, the physiological role of Ca2+ release via inositol 1,4,5-trisphosphate receptors (IP3Rs) from endoplasmic reticulum in regulating VSMC proliferation has not been well determined. METHODS AND RESULTS Both in vitro cell culture models and in vivo mouse models were generated to investigate the role of IP3Rs in regulating VSMC proliferation. Expression of all 3 IP3R subtypes was increased in cultured VSMCs upon platelet-derived growth factor-BB and FBS stimulation as well as in the left carotid artery undergoing intimal thickening after vascular occlusion. Genetic ablation of all 3 IP3R subtypes abolished endoplasmic reticulum Ca2+ release in cultured VSMCs, significantly reduced cell proliferation induced by platelet-derived growth factor-BB and FBS stimulation, and also decreased cell migration of VSMCs. Furthermore, smooth muscle-specific deletion of all IP3R subtypes in adult mice dramatically attenuated neointima formation induced by left carotid artery ligation, accompanied by significant decreases in cell proliferation and matrix metalloproteinase-9 expression in injured vessels. Mechanistically, IP3R-mediated Ca2+ release may activate cAMP response element-binding protein, a key player in controlling VSMC proliferation, via Ca2+/calmodulin-dependent protein kinase II and Akt. Loss of IP3Rs suppressed cAMP response element-binding protein phosphorylation at Ser133 in both cultured VSMCs and injured vessels, whereas application of Ca2+ permeable ionophore, ionomycin, can reverse cAMP response element-binding protein phosphorylation in IP3R triple knockout VSMCs. CONCLUSIONS Our results demonstrated an essential role of IP3R-mediated Ca2+ release from endoplasmic reticulum in regulating cAMP response element-binding protein activation, VSMC proliferation, and neointima formation in mouse arteries.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Becaplermin/pharmacology
- Becaplermin/metabolism
- Calcium/metabolism
- Calcium Signaling
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/metabolism
- Cyclic AMP Response Element-Binding Protein/genetics
- Disease Models, Animal
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum/pathology
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/pathology
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
Collapse
Affiliation(s)
- Fang Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Fei Zhang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Qingen Da
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Yu Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Huihua Yang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Hong Wang
- Central LaboratoryPeking University Shenzhen HospitalShenzhenChina
| | - Lingyun Zhao
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Qingsong Lin
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Zee Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Jie Liu
- Department of Pathophysiology, School of MedicineShenzhen UniversityShenzhenChina
| | - Mingming Ren
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Yan Wang
- Department of CardiologyQingdao Municipal HospitalQingdaoChina
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate SchoolPeking UniversityShenzhenChina
| |
Collapse
|
4
|
Yu M, Harper AR, Aguirre M, Pittman M, Tcheandjieu C, Amgalan D, Grace C, Goel A, Farrall M, Xiao K, Engreitz J, Pollard KS, Watkins H, Priest JR. Genetic Determinants of the Interventricular Septum Are Linked to Ventricular Septal Defects and Hypertrophic Cardiomyopathy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:207-215. [PMID: 37017090 PMCID: PMC10293084 DOI: 10.1161/circgen.122.003708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 01/06/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND A large proportion of genetic risk remains unexplained for structural heart disease involving the interventricular septum (IVS) including hypertrophic cardiomyopathy and ventricular septal defects. This study sought to develop a reproducible proxy of IVS structure from standard medical imaging, discover novel genetic determinants of IVS structure, and relate these loci to diseases of the IVS, hypertrophic cardiomyopathy, and ventricular septal defect. METHODS We estimated the cross-sectional area of the IVS from the 4-chamber view of cardiac magnetic resonance imaging in 32 219 individuals from the UK Biobank which was used as the basis of genome wide association studies and Mendelian randomization. RESULTS Measures of IVS cross-sectional area at diastole were a strong proxy for the 3-dimensional volume of the IVS (Pearson r=0.814, P=0.004), and correlated with anthropometric measures, blood pressure, and diagnostic codes related to cardiovascular physiology. Seven loci with clear genomic consequence and relevance to cardiovascular biology were uncovered by genome wide association studies, most notably a single nucleotide polymorphism in an intron of CDKN1A (rs2376620; β, 7.7 mm2 [95% CI, 5.8-11.0]; P=6.0×10-10), and a common inversion incorporating KANSL1 predicted to disrupt local chromatin structure (β, 8.4 mm2 [95% CI, 6.3-10.9]; P=4.2×10-14). Mendelian randomization suggested that inheritance of larger IVS cross-sectional area at diastole was strongly associated with hypertrophic cardiomyopathy risk (pIVW=4.6×10-10) while inheritance of smaller IVS cross-sectional area at diastole was associated with risk for ventricular septal defect (pIVW=0.007). CONCLUSIONS Automated estimates of cross-sectional area of the IVS supports discovery of novel loci related to cardiac development and Mendelian disease. Inheritance of genetic liability for either small or large IVS, appears to confer risk for ventricular septal defect or hypertrophic cardiomyopathy, respectively. These data suggest that a proportion of risk for structural and congenital heart disease can be localized to the common genetic determinants of size and shape of cardiovascular anatomy.
Collapse
Affiliation(s)
- Mengyao Yu
- Dept of Pediatrics, Division of Pediatric Cardiology, Division of Cardiovascular Medicine, Stanford Univ School of Medicine
- Stanford Cardiovascular Institute, Stanford Univ, Stanford, CA
| | - Andrew R. Harper
- Radcliffe Dept of Medicine, Univ of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Matthew Aguirre
- Dept of Pediatrics, Division of Pediatric Cardiology, Division of Cardiovascular Medicine, Stanford Univ School of Medicine
- Dept of Biomedical Data Science, Stanford Medical School, Stanford
| | - Maureen Pittman
- Univ of California, San Francisco, San Francisco
- Gladstone Institute of Data Science & Biotechnology, San Francisco
| | - Catherine Tcheandjieu
- Dept of Pediatrics, Division of Pediatric Cardiology, Division of Cardiovascular Medicine, Stanford Univ School of Medicine
- Stanford Cardiovascular Institute, Stanford Univ, Stanford, CA
- Dept of Medicine, Division of Cardiovascular Medicine, Stanford Univ School of Medicine
| | - Dulguun Amgalan
- Dept of Genetics, Stanford Univ, Stanford, CA
- Basic Sciences and Engineering Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital, Stanford, CA
| | - Christopher Grace
- Radcliffe Dept of Medicine, Univ of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford
| | - Anuj Goel
- Radcliffe Dept of Medicine, Univ of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford
| | - Martin Farrall
- Radcliffe Dept of Medicine, Univ of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford
| | - Ke Xiao
- College of Information & Computer Sciences at Univ of Massachusetts Amherst, Amherst, MA
| | - Jesse Engreitz
- Dept of Genetics, Stanford Univ, Stanford, CA
- Basic Sciences and Engineering Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital, Stanford, CA
| | - Katherine S. Pollard
- Univ of California, San Francisco, San Francisco
- Gladstone Institute of Data Science & Biotechnology, San Francisco
- Chan-Zuckerberg Biohub
| | - Hugh Watkins
- Radcliffe Dept of Medicine, Univ of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford
| | - James R. Priest
- Dept of Pediatrics, Division of Pediatric Cardiology, Division of Cardiovascular Medicine, Stanford Univ School of Medicine
- Stanford Cardiovascular Institute, Stanford Univ, Stanford, CA
- Chan-Zuckerberg Biohub
- Current affiliation: Tenaya Therapeutics, South San Francisco, CA
| |
Collapse
|
5
|
Du X, Liu Y, He X, Tao L, Fang M, Chu M. Uterus proliferative period ceRNA network of Yunshang black goat reveals candidate genes on different kidding number trait. Front Endocrinol (Lausanne) 2023; 14:1165409. [PMID: 37251683 PMCID: PMC10213787 DOI: 10.3389/fendo.2023.1165409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Pregnancy loss that occurs in the uterus is an important and widespread problem in humans and farm animals and is also a key factor affecting the fecundity of livestock. Understanding the differences in the fecundity of goats may be helpful in guiding the breeding of goats with high fecundity. In this study, we performed RNA sequencing and bioinformatics analysis to study the uterus of Yunshang black goats with high and low fecundity in the proliferative period. We identified mRNAs, long non-coding RNAs (lncRNAs), and microRNAs (miRNAs) by analyzing the uterine transcriptomes. The target genes of the identified miRNAs and lncRNAs were predicted, and miRNA-mRNA interaction and competitive endogenous RNA (ceRNA) networks were constructed. By comparisons between low- and high-fecundity groups, we identified 1,674 differentially expressed mRNAs (914 were upregulated, and 760 were downregulated), 288 differentially expressed lncRNAs (149 were upregulated, and 139 were downregulated), and 17 differentially expressed miRNAs (4 were upregulated, and 13 were downregulated). In addition, 49 miRNA-mRNA pairs and 45 miRNA-lncRNA pairs were predicted in the interaction networks. We successfully constructed a ceRNA interaction network with 108 edges that contained 19 miRNAs, 11 mRNAs, and 73 lncRNAs. Five candidate genes (PLEKHA7, FAT2, FN1, SYK, and ITPR2) that were annotated as cell adhesion or calcium membrane channel protein were identified. Our results provide the overall expression profiles of mRNAs, lncRNAs, and miRNAs in the goat uterus during the proliferative period and are a valuable reference for studies into the mechanisms associated with the high fecundity, which may be helpful to guide goat to reduce pregnancy loss.
Collapse
Affiliation(s)
- Xiaolong Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, Ministry of Agriculture and Rural Affairs (MARA) PRC Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yufang Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lin Tao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, Ministry of Agriculture and Rural Affairs (MARA) PRC Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
6
|
Genetic and functional analyses of TBX4 reveal novel mechanisms underlying pulmonary arterial hypertension. J Mol Cell Cardiol 2022; 171:105-116. [PMID: 35914404 DOI: 10.1016/j.yjmcc.2022.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a fatal disease, with approximately 10% of cases associated with genetic variants. Recent genetic studies have reported pathogenic variants in the TBX4 gene in patients with PAH, especially in patients with childhood-onset of the disease, but the pathogenesis of PAH caused by TBX4 variant has not been fully uncovered. METHODS We analysed the TBX4 gene in 75 Japanese patients with sporadic or familial PAH using a PCR-based bidirectional sequencing method. Detected variants were evaluated using in silico analyses as well as in vitro analyses including luciferase assay, immunocytochemistry and chromatin immunoprecipitation (ChIP) whether they have altered function. We also analysed the function of TBX4 using mouse embryonic lung explants with inhibition of Tbx4 expression. RESULTS Putative pathogenic variants were detected in three cases (4.0%). Our in vitro functional analyses revealed that TBX4 directly regulates the transcriptional activity of fibroblast growth factor 10 (FGF10), whereas the identified TBX4 variant proteins failed to activate the FGF10 gene because of disruption of nuclear localisation signal or poor DNA-binding affinity. Furthermore, ex vivo inhibition of Tbx4 resulted in insufficiency of lung morphogenesis along with specific downregulation of Tie2 and Kruppel-like factor 4 expression. CONCLUSION Our results implicate variants in TBX4 as a genetic cause of PAH in a subset of the Japanese population. Variants in TBX4 may lead to PAH through insufficient lung morphogenesis by disrupting the TBX4-mediated direct regulation of FGF10 signalling and pulmonary vascular endothelial dysfunction involving PAH-related molecules.
Collapse
|
7
|
Young MP, Schug ZT, Booth DM, Yule DI, Mikoshiba K, Hajnóczky G, Joseph SK. Metabolic adaptation to the chronic loss of Ca 2+ signaling induced by KO of IP 3 receptors or the mitochondrial Ca 2+ uniporter. J Biol Chem 2022; 298:101436. [PMID: 34801549 PMCID: PMC8672050 DOI: 10.1016/j.jbc.2021.101436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/04/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023] Open
Abstract
Calcium signaling is essential for regulating many biological processes. Endoplasmic reticulum inositol trisphosphate receptors (IP3Rs) and the mitochondrial Ca2+ uniporter (MCU) are key proteins that regulate intracellular Ca2+ concentration. Mitochondrial Ca2+ accumulation activates Ca2+-sensitive dehydrogenases of the tricarboxylic acid (TCA) cycle that maintain the biosynthetic and bioenergetic needs of both normal and cancer cells. However, the interplay between calcium signaling and metabolism is not well understood. In this study, we used human cancer cell lines (HEK293 and HeLa) with stable KOs of all three IP3R isoforms (triple KO [TKO]) or MCU to examine metabolic and bioenergetic responses to the chronic loss of cytosolic and/or mitochondrial Ca2+ signaling. Our results show that TKO cells (exhibiting total loss of Ca2+ signaling) are viable, displaying a lower proliferation and oxygen consumption rate, with no significant changes in ATP levels, even when made to rely solely on the TCA cycle for energy production. MCU KO cells also maintained normal ATP levels but showed increased proliferation, oxygen consumption, and metabolism of both glucose and glutamine. However, MCU KO cells were unable to maintain ATP levels and died when relying solely on the TCA cycle for energy. We conclude that constitutive Ca2+ signaling is dispensable for the bioenergetic needs of both IP3R TKO and MCU KO human cancer cells, likely because of adequate basal glycolytic and TCA cycle flux. However, in MCU KO cells, the higher energy expenditure associated with increased proliferation and oxygen consumption makes these cells more prone to bioenergetic failure under conditions of metabolic stress.
Collapse
Affiliation(s)
- Michael P Young
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Zachary T Schug
- Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - David M Booth
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - David I Yule
- Department of Pharmacology & Physiology, University of Rochester, Rochester, New York, USA
| | - Katsuhiko Mikoshiba
- Shanghai Institute of Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China; Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Japan
| | - György Hajnóczky
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Suresh K Joseph
- Department of Pathology, MitoCare Center, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
8
|
Kodo K, Uchida K, Yamagishi H. Genetic and Cellular Interaction During Cardiovascular Development Implicated in Congenital Heart Diseases. Front Cardiovasc Med 2021; 8:653244. [PMID: 33796576 PMCID: PMC8007765 DOI: 10.3389/fcvm.2021.653244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/22/2021] [Indexed: 12/21/2022] Open
Abstract
Congenital heart disease (CHD) is the most common life-threatening congenital anomaly. CHD occurs due to defects in cardiovascular development, and the majority of CHDs are caused by a multifactorial inheritance mechanism, which refers to the interaction between genetic and environmental factors. During embryogenesis, the cardiovascular system is derived from at least four distinct cell lineages: the first heart field, second heart field, cardiac neural crest, and proepicardial organ. Understanding the genes involved in each lineage is essential to uncover the genomic architecture of CHD. Therefore, we provide an overview of recent research progress using animal models and mutation analyses to better understand the molecular mechanisms and pathways linking cardiovascular development and CHD. For example, we highlight our recent work on genes encoding three isoforms of inositol 1,4,5-trisphosphate receptors (IP3R1, 2, and 3) that regulate various vital and developmental processes, which have genetic redundancy during cardiovascular development. Specifically, IP3R1 and 2 have redundant roles in the atrioventricular cushion derived from the first heart field lineage, whereas IP3R1 and 3 exhibit redundancy in the right ventricle and the outflow tract derived from the second heart field lineage, respectively. Moreover, 22q11.2 deletion syndrome (22q11DS) is highly associated with CHD involving the outflow tract, characterized by defects of the cardiac neural crest lineage. However, our studies have shown that TBX1, a major genetic determinant of 22q11DS, was not expressed in the cardiac neural crest but rather in the second heart field, suggesting the importance of the cellular interaction between the cardiac neural crest and the second heart field. Comprehensive genetic analysis using the Japanese genome bank of CHD and mouse models revealed that a molecular regulatory network involving GATA6, FOXC1/2, TBX1, SEMA3C, and FGF8 was essential for reciprocal signaling between the cardiac neural crest and the second heart field during cardiovascular development. Elucidation of the genomic architecture of CHD using induced pluripotent stem cells and next-generation sequencing technology, in addition to genetically modified animal models and human mutation analyses, would facilitate the development of regenerative medicine and/or preventive medicine for CHD in the near future.
Collapse
Affiliation(s)
- Kazuki Kodo
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Uchida
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Yamagishi
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Izarzugaza JMG, Ellesøe SG, Doganli C, Ehlers NS, Dalgaard MD, Audain E, Dombrowsky G, Banasik K, Sifrim A, Wilsdon A, Thienpont B, Breckpot J, Gewillig M, Brook JD, Hitz MP, Larsen LA, Brunak S. Systems genetics analysis identifies calcium-signaling defects as novel cause of congenital heart disease. Genome Med 2020; 12:76. [PMID: 32859249 PMCID: PMC7453558 DOI: 10.1186/s13073-020-00772-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 08/07/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) occurs in almost 1% of newborn children and is considered a multifactorial disorder. CHD may segregate in families due to significant contribution of genetic factors in the disease etiology. The aim of the study was to identify pathophysiological mechanisms in families segregating CHD. METHODS We used whole exome sequencing to identify rare genetic variants in ninety consenting participants from 32 Danish families with recurrent CHD. We applied a systems biology approach to identify developmental mechanisms influenced by accumulation of rare variants. We used an independent cohort of 714 CHD cases and 4922 controls for replication and performed functional investigations using zebrafish as in vivo model. RESULTS We identified 1785 genes, in which rare alleles were shared between affected individuals within a family. These genes were enriched for known cardiac developmental genes, and 218 of these genes were mutated in more than one family. Our analysis revealed a functional cluster, enriched for proteins with a known participation in calcium signaling. Replication in an independent cohort confirmed increased mutation burden of calcium-signaling genes in CHD patients. Functional investigation of zebrafish orthologues of ITPR1, PLCB2, and ADCY2 verified a role in cardiac development and suggests a combinatorial effect of inactivation of these genes. CONCLUSIONS The study identifies abnormal calcium signaling as a novel pathophysiological mechanism in human CHD and confirms the complex genetic architecture underlying CHD.
Collapse
Affiliation(s)
- Jose M G Izarzugaza
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
| | - Sabrina G Ellesøe
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Canan Doganli
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Natasja Spring Ehlers
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
| | - Marlene D Dalgaard
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
- DTU Multi Assay Core (DMAC), Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark
| | - Enrique Audain
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Gregor Dombrowsky
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Alejandro Sifrim
- Wellcome Trust Sanger Institute, Cambridge, UK
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anna Wilsdon
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Bernard Thienpont
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jeroen Breckpot
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Marc Gewillig
- Pediatric Cardiology Unit, University Hospitals Leuven, Leuven, Belgium
| | - J David Brook
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Marc-Phillip Hitz
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
- Wellcome Trust Sanger Institute, Cambridge, UK
- Institute of Human Genetics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Lars A Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark.
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
10
|
Tekola-Ayele F, Zhang C, Wu J, Grantz KL, Rahman ML, Shrestha D, Ouidir M, Workalemahu T, Tsai MY. Trans-ethnic meta-analysis of genome-wide association studies identifies maternal ITPR1 as a novel locus influencing fetal growth during sensitive periods in pregnancy. PLoS Genet 2020; 16:e1008747. [PMID: 32407400 PMCID: PMC7252673 DOI: 10.1371/journal.pgen.1008747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 05/27/2020] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Abnormal fetal growth is a risk factor for infant morbidity and mortality and is associated with cardiometabolic diseases in adults. Genetic influences on fetal growth can vary at different gestation times, but genome-wide association studies have been limited to birthweight. We performed trans-ethnic genome-wide meta-analyses and fine mapping to identify maternal genetic loci associated with fetal weight estimates obtained from ultrasound measures taken during pregnancy. Data included 1,849 pregnant women from four race/ethnic groups recruited through the NICHD Fetal Growth Studies. We identified a novel genome-wide significant association of rs746039 [G] (ITPR1) with reduced fetal weight from 24 to 33 weeks gestation (P<5x10-8; log10BF>6). Additional tests revealed that the SNP was associated with head circumference (P = 4.85x10-8), but not with abdominal circumference or humerus/femur lengths. Conditional analysis in an independent sample of mother-offspring pairs replicated the findings and showed that the effect was more likely maternal but not fetal. Trans-ethnic approaches successfully narrowed down the haplotype block that contained the 99% credible set of SNPs associated with head circumference. We further demonstrated that decreased placental expression of ITPR1 was correlated with increased placental epigenetic age acceleration, a risk factor for reduced fetal growth, among male fetuses (r = -0.4, P = 0.01). Finally, genetic risk score composed of known maternal SNPs implicated in birthweight among Europeans was associated with fetal weight from mid-gestation onwards among Whites only. The present study sheds new light on the role of common maternal genetic variants in the inositol receptor signaling pathway on fetal growth from late second trimester to early third trimester. Clinical Trial Registration: ClinicalTrials.gov, NCT00912132.
Collapse
Affiliation(s)
- Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cuilin Zhang
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jing Wu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katherine L. Grantz
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mohammad L. Rahman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Population Medicine and Harvard Pilgrim Healthcare Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Deepika Shrestha
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marion Ouidir
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tsegaselassie Workalemahu
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
11
|
Yang F, Huang L, Tso A, Wang H, Cui L, Lin L, Wang X, Ren M, Fang X, Liu J, Han Z, Chen J, Ouyang K. Inositol 1,4,5-trisphosphate receptors are essential for fetal-maternal connection and embryo viability. PLoS Genet 2020; 16:e1008739. [PMID: 32320395 PMCID: PMC7176088 DOI: 10.1371/journal.pgen.1008739] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/25/2020] [Indexed: 01/28/2023] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are a family of intracellular Ca2+ release channels located on the ER membrane, which in mammals consist of 3 different subtypes (IP3R1, IP3R2, and IP3R3) encoded by 3 genes, Itpr1, Itpr2, and Itpr3, respectively. Studies utilizing genetic knockout mouse models have demonstrated that IP3Rs are essential for embryonic survival in a redundant manner. Deletion of both IP3R1 and IP3R2 has been shown to cause cardiovascular defects and embryonic lethality. However, it remains unknown which cell types account for the cardiovascular defects in IP3R1 and IP3R2 double knockout (DKO) mice. In this study, we generated conditional IP3R1 and IP3R2 knockout mouse models with both genes deleted in specific cardiovascular cell lineages. Our results revealed that deletion of IP3R1 and IP3R2 in cardiomyocytes by TnT-Cre, in endothelial / hematopoietic cells by Tie2-Cre and Flk1-Cre, or in early precursors of the cardiovascular lineages by Mesp1-Cre, resulted in no phenotypes. This demonstrated that deletion of both IP3R genes in cardiovascular cell lineages cannot account for the cardiovascular defects and embryonic lethality observed in DKO mice. We then revisited and performed more detailed phenotypic analysis in DKO embryos, and found that DKO embryos developed cardiovascular defects including reduced size of aortas, enlarged cardiac chambers, as well as growth retardation at embryonic day (E) 9.5, but in varied degrees of severity. Interestingly, we also observed allantoic-placental defects including reduced sizes of umbilical vessels and reduced depth of placental labyrinth in DKO embryos, which could occur independently from other phenotypes in DKO embryos even without obvious growth retardation. Furthermore, deletion of both IP3R1 and IP3R2 by the epiblast-specific Meox2-Cre, which targets all the fetal tissues and extraembryonic mesoderm but not extraembryonic trophoblast cells, also resulted in embryonic lethality and similar allantoic-placental defects. Taken together, our results demonstrated that IP3R1 and IP3R2 play an essential and redundant role in maintaining the integrity of fetal-maternal connection and embryonic viability.
Collapse
Affiliation(s)
- Feili Yang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Alexandria Tso
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, CA, United States of America
| | - Hong Wang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Li Cui
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, CA, United States of America
| | - Lizhu Lin
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, CA, United States of America
| | - Xiaohong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mingming Ren
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xi Fang
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, CA, United States of America
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- * E-mail: (ZH); (JC); (KO)
| | - Ju Chen
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, CA, United States of America
- * E-mail: (ZH); (JC); (KO)
| | - Kunfu Ouyang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
- * E-mail: (ZH); (JC); (KO)
| |
Collapse
|
12
|
Gambardella J, Lombardi A, Morelli MB, Ferrara J, Santulli G. Inositol 1,4,5-Trisphosphate Receptors in Human Disease: A Comprehensive Update. J Clin Med 2020; 9:1096. [PMID: 32290556 PMCID: PMC7231134 DOI: 10.3390/jcm9041096] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/22/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (ITPRs) are intracellular calcium release channels located on the endoplasmic reticulum of virtually every cell. Herein, we are reporting an updated systematic summary of the current knowledge on the functional role of ITPRs in human disorders. Specifically, we are describing the involvement of its loss-of-function and gain-of-function mutations in the pathogenesis of neurological, immunological, cardiovascular, and neoplastic human disease. Recent results from genome-wide association studies are also discussed.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY 10461, USA; (J.G.); (A.L.); (M.B.M.); (J.F.)
- International Translational Research and Medical Education Consortium (ITME), 80100 Naples, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Angela Lombardi
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY 10461, USA; (J.G.); (A.L.); (M.B.M.); (J.F.)
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Marco Bruno Morelli
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY 10461, USA; (J.G.); (A.L.); (M.B.M.); (J.F.)
- Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - John Ferrara
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY 10461, USA; (J.G.); (A.L.); (M.B.M.); (J.F.)
| | - Gaetano Santulli
- Department of Medicine, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York, NY 10461, USA; (J.G.); (A.L.); (M.B.M.); (J.F.)
- International Translational Research and Medical Education Consortium (ITME), 80100 Naples, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
13
|
Guo X, Zhang W, Li M, Gao P, Hei W, He Z, Wu Y, Liu J, Cai C, Li B, Cao G. Transcriptome profile of skeletal muscle at different developmental stages in Large White and Mashen pigs. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2019-0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
From the perspectives of promoting individual growth and development, increasing pork yield, and improving feed utilization, it is desirable to screen candidate genes underlying pig muscle growth and regulation. In this study, we investigated transcriptome differences at 1, 90, and 180 d of age in Large White and Mashen pigs, characterized differentially expressed genes (DEGs), and screened candidate genes affecting skeletal muscle growth and development. RNA-seq was applied to analyze the transcriptome of the longissimus dorsi (LD) in the two breeds. In LD samples from the two breeds at three growth stages, 7215, 6332, 237, 3935, 3404, and 846 DEGs were obtained for L01 vs. L90, L01 vs. L180, L90 vs. L180, MS01 vs. MS90, MS01 vs. MS180, and MS90 vs. MS180, respectively. Significant tendencies in DEG expression could be grouped into eight profiles. Based on the functional analysis of DEGs, 16 candidate genes related to skeletal muscle growth and development were identified, including PCK2, GNAS, ADCY2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, PHKA1, PHKA2, PHKG1, PHKG2, ITPR3, IGF1R, FGFR4, FGF1, and FGF18. The results of this study thus provide a theoretical basis for the mechanisms and candidate genes underlying skeletal muscle development in pigs.
Collapse
Affiliation(s)
- Xiaohong Guo
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Wanfeng Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Meng Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Pengfei Gao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Wei Hei
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Zhiqiang He
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Yiqi Wu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Juan Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Chunbo Cai
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Bugao Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Guoqing Cao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| |
Collapse
|
14
|
Ishizaki-Asami R, Uchida K, Tsuchihashi T, Shibata A, Kodo K, Emoto K, Mikoshiba K, Takahashi T, Yamagishi H. Inositol 1,4,5-trisphosphate receptor 2 as a novel marker of vasculature to delineate processes of cardiopulmonary development. Dev Biol 2019; 458:237-245. [PMID: 31758944 DOI: 10.1016/j.ydbio.2019.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/14/2019] [Accepted: 11/19/2019] [Indexed: 11/26/2022]
Abstract
Congenital heart diseases (CHDs) involving the outflow tract (OFT), such as persistent truncus arteriosus (PTA), lead to mortality and morbidity with implications not only in the heart, but also in the pulmonary vasculature. The mechanisms of pulmonary artery (PA) development and the etiologies underlying PA disorders associated with CHD remain poorly understood partly because of a specific marker for PA development is nonexistent. The three subtypes of inositol 1,4,5-trisphosphate receptors (IP3R1, 2, and 3) are intracellular Ca2+ channels that are essential for many tissues and organs. We discovered that IP3R2 was expressed in the vasculature and heart during development using transgenic mice, in which a LacZ marker gene was knocked into the IP3R2 locus. Whole-mount and section LacZ staining showed that IP3R2-LacZ-positive cells were detectable exclusively in the smooth muscle cells, or tunica media, of PA, merging into αSMA-positive cells during development. Furthermore, our analyses suggested that IP3R2-LacZ positive PA smooth muscle layers gradually elongate from the central PA to the peripheral PAs from E13.5 to E18.5, supporting the distal angiogenesis theory for the development of PA, whereas IP3R2-LacZ was rarely expressed in smooth muscle cells in the pulmonary trunk. Crossing IP3R-LacZ mice with mice hypomorphic for Tbx1 alleles revealed that PTA of Tbx1 mutants may result from agenesis or hypoplasia of the pulmonary trunk; thus, the left and right central to peripheral PAs connect directly to the dorsal side of the truncus arteriosus in these mutants. Additionally, we found hypercellular interstitial mesenchyme and delayed maturation of the lung endoderm in the Tbx1 mutant lungs. Our study identifies IP3R2 as a novel marker for clear visualization of PA during development and can be utilized for studying cardiopulmonary development and disease.
Collapse
Affiliation(s)
- Reina Ishizaki-Asami
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Keiko Uchida
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Health Center, Keio University, 4-1-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8521, Japan.
| | - Takatoshi Tsuchihashi
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Pediatrics, Kawasaki Municipal Hospital, 12-1 Shinkawadōri, Kawasaki-ku, Kawasaki, Kanagawa, 210-0013, Japan
| | - Akimichi Shibata
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Pediatrics, Japanese Red Cross Ashikaga Hospital, 284-1 Yobe-cho, Ashikaga, Tochigi, 326-0843, Japan
| | - Kazuki Kodo
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Katsura Emoto
- Division of Diagnostic Pathology, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Katsuhiko Mikoshiba
- SIAIS (Shanghai Institute for Advanced Immunochemical Studies), ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China; Toho University, Faculty of Science, Miyama 2-2-1, Funabashi, Chiba, 274-8510, Japan; Laboratory for Developmental Neurobiology, Center for Brain Sciences, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takao Takahashi
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Yamagishi
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
15
|
Chen B, Qi CY, Chen L, Dai MJ, Miao YY, Chen R, Wei WE, Yang S, Wang HL, Duan XG, Gong MW, Wang Y, Xue ZF. A C1976Y missense mutation in the mouse Ip3r1 gene leads to short-term mydriasis and unfolded protein response in the iris constrictor muscles. Exp Anim 2019; 69:45-53. [PMID: 31391379 PMCID: PMC7004804 DOI: 10.1538/expanim.19-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Ip3r1 encodes an inositol 1,4,5-trisphosphate-responsive calcium
channel. Mutations in the IP3R1 gene in humans may cause Gillespie
syndrome (GS) typically presents as fixed dilated pupils in affected infants, which was
referred to as iris hypoplasia. However, there is no report of mice with
Ip3r1 heterozygous mutations showing dilated pupils. Here, we report a
new Ip3r1 allele with short-term dilated pupil phenotype derived from an
N-ethyl-N-nitrosourea (ENU) mutagenesis screen. This allele carries a G5927A transition
mutation in Ip3r1 gene (NM_010585), which is predicted to result in a
C1976Y amino acid change in the open reading frame of IP3R1 (NP_034715). We named this
novel Ip3r1 allele Ip3r1C1976Y. Histology and
pharmacological tests show that the dilated pupil phenotype is a mydriasis caused by the
functional defect in the iris constrictor muscles in
Ip3r1C1976Y. The dilated pupil phenotype in
Ip3r1C1976Y was referred to as mydriasis and excluding
iris hypoplasia. IHC analysis revealed increased expression of BIP protein, the master
regulator of unfolded protein response (UPR) signaling, in
Ip3r1C1976Y mice that did not recover. This study is the
first report of an Ip3r1 mutation being associated with the mydriasis
phenotype. Ip3r1C1976Y mice represent a self-healing model
that may be used to study the therapeutic approach for Ip3r1-related
diseases.
Collapse
Affiliation(s)
- Bing Chen
- Institute of Comparative Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China.,College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Chong-Yang Qi
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Li Chen
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Meng-Jun Dai
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Ya-You Miao
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Rui Chen
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Wan-E Wei
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Shun Yang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Hong-Ling Wang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Xiao-Ge Duan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Min-Wei Gong
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Yi Wang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| | - Zheng-Feng Xue
- Institute of Comparative Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China.,College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 12 Wenhui East Road, Yangzhou, Jiangsu Province 225009, P.R.China
| |
Collapse
|
16
|
Wang H, Jing R, Trexler C, Li Y, Tang H, Pan Z, Zhu S, Zhao B, Fang X, Liu J, Chen J, Ouyang K. Deletion of IP 3R1 by Pdgfrb-Cre in mice results in intestinal pseudo-obstruction and lethality. J Gastroenterol 2019; 54:407-418. [PMID: 30382364 PMCID: PMC8109192 DOI: 10.1007/s00535-018-1522-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/17/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Inositol 1,4,5-trisphosphate receptors (IP3Rs) are a family of intracellular Ca2+ release channels located on the membrane of endoplasmic reticulum, which have been shown to play critical roles in various cellular and physiological functions. However, their function in regulating gastrointestinal (GI) tract motility in vivo remains unknown. Here, we investigated the physiological function of IP3R1 in the GI tract using genetically engineered mouse models. METHODS Pdgfrb-Cre mice were bred with homozygous Itpr1 floxed (Itpr1f/f) mice to generate conditional IP3R1 knockout (pcR1KO) mice. Cell lineage tracing was used to determine where Pdgfrb-Cre-mediated gene deletion occurred in the GI tract. Isometric tension recording was used to measure the effects of IP3R1 deletion on muscle contraction. RESULTS In the mouse GI tract, Itpr1 gene deletion by Pdgfrb-Cre occurred in smooth muscle cells, enteric neurons, and interstitial cells of Cajal. pcR1KO mice developed impaired GI motility, with prolonged whole-gut transit time and abdominal distention. pcR1KO mice also exhibited lethality as early as 8 weeks of age and 50% of pcR1KO mice were dead by 40 weeks after birth. The frequency of spontaneous contractions in colonic circular muscles was dramatically decreased and the amplitude of spontaneous contractions was increased in pcR1KO mice. Deletion of IP3R1 in the GI tract also reduced the contractile response to the muscarinic agonist, carbachol, as well as to electrical field stimulation. However, KCl-induced contraction and expression of smooth muscle-specific contractile genes were not significantly altered in pcR1KO mice. CONCLUSIONS Here, we provided a novel mouse model for impaired GI motility and demonstrated that IP3R1 plays a critical role in regulating physiological function of GI tract in vivo.
Collapse
Affiliation(s)
- Hong Wang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Ran Jing
- Xiangya Hospital, Central South University, Changsha 410011, China
| | - Christa Trexler
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yali Li
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Huayuan Tang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Zhixiang Pan
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Siting Zhu
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Beili Zhao
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xi Fang
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Ju Chen
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
17
|
Type 2 inositol 1,4,5-trisphosphate receptor inhibits the progression of pulmonary arterial hypertension via calcium signaling and apoptosis. Heart Vessels 2018; 34:724-734. [PMID: 30460575 DOI: 10.1007/s00380-018-1304-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease associated with vasoconstriction and remodeling. Intracellular Ca2+ signaling regulates the contraction of pulmonary arteries and the proliferation of pulmonary arterial smooth muscle cells (PASMCs); however, it is not clear which molecules related to Ca2+ signaling contribute to the progression of PAH. In this study, we found the specific expression of type 2 inositol 1,4,5-trisphosphate receptor (IP3R2), which is an intracellular Ca2+ release channel, on the sarco/endoplasmic reticulum in mouse PASMCs, and demonstrated its inhibitory role in the progression of PAH using a chronic hypoxia-induced PAH mouse model. After chronic hypoxia exposure, IP3R2-/- mice exhibited the significant aggravation of PAH, as determined by echocardiography and right ventricular hypertrophy, with significantly greater medial wall thickness by immunohistochemistry than that of wild-type mice. In IP3R2-/- murine PASMCs with chronic hypoxia, a TUNEL assay revealed the significant suppression of apoptosis, whereas there was no significant change in proliferation. Thapsigargin-induced store-operated Ca2+ entry (SOCE) was significantly enhanced in IP3R2-/- PASMCs in both normoxia and hypoxia based on in vitro fluorescent Ca2+ imaging. Furthermore, the enhancement of SOCE in IP3R2-/- PASMCs was remarkably suppressed by the addition of DPB162-AE, an inhibitor of the stromal-interacting molecule (STIM)-Orai complex which is about 100 times more potent than 2-APB. Our results indicate that IP3R2 may inhibit the progression of PAH by promoting apoptosis and inhibiting SOCE via the STIM-Orai pathway in PASMCs. These findings suggest a previously undetermined role of IP3R in the development of PAH and may contribute to the development of targeted therapies.
Collapse
|
18
|
Wang YJ, Huang J, Liu W, Kou X, Tang H, Wang H, Yu X, Gao S, Ouyang K, Yang HT. IP3R-mediated Ca2+ signals govern hematopoietic and cardiac divergence of Flk1+ cells via the calcineurin-NFATc3-Etv2 pathway. J Mol Cell Biol 2018; 9:274-288. [PMID: 28419336 DOI: 10.1093/jmcb/mjx014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 04/10/2017] [Indexed: 12/30/2022] Open
Abstract
Ca2+ signals participate in various cellular processes with spatial and temporal dynamics, among which, inositol 1,4,5-trisphosphate receptors (IP3Rs)-mediated Ca2+ signals are essential for early development. However, the underlying mechanisms of IP3R-regulated cell fate decision remain largely unknown. Here we report that IP3Rs are required for the hematopoietic and cardiac fate divergence of mouse embryonic stem cells (mESCs). Deletion of IP3Rs (IP3R-tKO) reduced Flk1+/PDGFRα- hematopoietic mesoderm, c-Kit+/CD41+ hematopoietic progenitor cell population, and the colony-forming unit activity, but increased cardiac progenitor markers as well as cardiomyocytes. Concomitantly, the expression of a key regulator of hematopoiesis, Etv2, was reduced in IP3R-tKO cells, which could be rescued by the activation of Ca2+ signals and calcineurin or overexpression of constitutively active form of NFATc3. Furthermore, IP3R-tKO impaired specific targeting of Etv2 by NFATc3 via its evolutionarily conserved cis-element in differentiating ESCs. Importantly, the activation of Ca2+-calcineurin-NFAT pathway reversed the phenotype of IP3R-tKO cells. These findings reveal an unrecognized governing role of IP3Rs in hematopoietic and cardiac fate commitment via IP3Rs-Ca2+-calcineurin-NFATc3-Etv2 pathway.
Collapse
Affiliation(s)
- Yi-Jie Wang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Jijun Huang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Wenqiang Liu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaochen Kou
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Huayuan Tang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Hong Wang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiujian Yu
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kunfu Ouyang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Huang-Tian Yang
- Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
19
|
Tang H, Wang H, Lin Q, Fan F, Zhang F, Peng X, Fang X, Liu J, Ouyang K. Loss of IP3 Receptor–Mediated Ca2+ Release in Mouse B Cells Results in Abnormal B Cell Development and Function. THE JOURNAL OF IMMUNOLOGY 2017; 199:570-580. [DOI: 10.4049/jimmunol.1700109] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/17/2017] [Indexed: 12/16/2022]
|
20
|
Lin Q, Zhao G, Fang X, Peng X, Tang H, Wang H, Jing R, Liu J, Lederer WJ, Chen J, Ouyang K. IP 3 receptors regulate vascular smooth muscle contractility and hypertension. JCI Insight 2016; 1:e89402. [PMID: 27777977 DOI: 10.1172/jci.insight.89402] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Inositol 1, 4, 5-trisphosphate receptor-mediated (IP3R-mediated) calcium (Ca2+) release has been proposed to play an important role in regulating vascular smooth muscle cell (VSMC) contraction for decades. However, whether and how IP3R regulates blood pressure in vivo remains unclear. To address these questions, we have generated a smooth muscle-specific IP3R triple-knockout (smTKO) mouse model using a tamoxifen-inducible system. In this study, the role of IP3R-mediated Ca2+ release in adult VSMCs on aortic vascular contractility and blood pressure was assessed following tamoxifen induction. We demonstrated that deletion of IP3Rs significantly reduced aortic contractile responses to vasoconstrictors, including phenylephrine, U46619, serotonin, and endothelin 1. Deletion of IP3Rs also dramatically reduced the phosphorylation of MLC20 and MYPT1 induced by U46619. Furthermore, although the basal blood pressure of smTKO mice remained similar to that of wild-type controls, the increase in systolic blood pressure upon chronic infusion of angiotensin II was significantly attenuated in smTKO mice. Taken together, our results demonstrate an important role for IP3R-mediated Ca2+ release in VSMCs in regulating vascular contractility and hypertension.
Collapse
Affiliation(s)
- Qingsong Lin
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Guiling Zhao
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xi Fang
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, California, USA
| | - Xiaohong Peng
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Huayuan Tang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Hong Wang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ran Jing
- Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ju Chen
- University of California San Diego, School of Medicine, Department of Medicine, La Jolla, California, USA
| | - Kunfu Ouyang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
21
|
Type 1 and 3 inositol trisphosphate receptors are required for extra-embryonic vascular development. Dev Biol 2016; 418:89-97. [DOI: 10.1016/j.ydbio.2016.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 08/07/2016] [Indexed: 11/17/2022]
|
22
|
Huang J, Zhang M, Zhang P, Liang H, Ouyang K, Yang HT. Coupling switch of P2Y-IP3 receptors mediates differential Ca(2+) signaling in human embryonic stem cells and derived cardiovascular progenitor cells. Purinergic Signal 2016; 12:465-78. [PMID: 27098757 DOI: 10.1007/s11302-016-9512-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/04/2016] [Indexed: 12/18/2022] Open
Abstract
Purinergic signaling mediated by P2 receptors (P2Rs) plays important roles in embryonic and stem cell development. However, how it mediates Ca(2+) signals in human embryonic stem cells (hESCs) and derived cardiovascular progenitor cells (CVPCs) remains unclear. Here, we aimed to determine the role of P2Rs in mediating Ca(2+) mobilizations of these cells. hESCs were induced to differentiate into CVPCs by our recently established methods. Gene expression of P2Rs and inositol 1,4,5-trisphosphate receptors (IP3Rs) was analyzed by quantitative/RT-PCR. IP3R3 knockdown (KD) or IP3R2 knockout (KO) hESCs were established by shRNA- or TALEN-mediated gene manipulations, respectively. Confocal imaging revealed that Ca(2+) responses in CVPCs to ATP and UTP were more sensitive and stronger than those in hESCs. Consistently, the gene expression levels of most P2YRs except P2Y1 were increased in CVPCs. Suramin or PPADS blocked ATP-induced Ca(2+) transients in hESCs but only partially inhibited those in CVPCs. Moreover, the P2Y1 receptor-specific antagonist MRS2279 abolished most ATP-induced Ca(2+) signals in hESCs but not in CVPCs. P2Y1 receptor-specific agonist MRS2365 induced Ca(2+) transients only in hESCs but not in CVPCs. Furthermore, IP3R2KO but not IP3R3KD decreased the proportion of hESCs responding to MRS2365. In contrast, both IP3R2 and IP3R3 contributed to UTP-induced Ca(2+) responses while ATP-induced Ca(2+) responses were more dependent on IP3R2 in the CVPCs. In conclusion, a predominant role of P2Y1 receptors in hESCs and a transition of P2Y-IP3R coupling in derived CVPCs are responsible for the differential Ca(2+) mobilization between these cells.
Collapse
Affiliation(s)
- Jijun Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.,Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Min Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China
| | - Peng Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China
| | - He Liang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Kunfu Ouyang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Huang-Tian Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China. .,Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China. .,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
23
|
Barnes RM, Harris IS, Jaehnig EJ, Sauls K, Sinha T, Rojas A, Schachterle W, McCulley DJ, Norris RA, Black BL. MEF2C regulates outflow tract alignment and transcriptional control of Tdgf1. Development 2016; 143:774-9. [PMID: 26811383 DOI: 10.1242/dev.126383] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 01/19/2016] [Indexed: 01/24/2023]
Abstract
Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.
Collapse
Affiliation(s)
- Ralston M Barnes
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Ian S Harris
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric J Jaehnig
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Kimberly Sauls
- Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Anabel Rojas
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - William Schachterle
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - David J McCulley
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Russell A Norris
- Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Vervloessem T, Yule DI, Bultynck G, Parys JB. The type 2 inositol 1,4,5-trisphosphate receptor, emerging functions for an intriguing Ca²⁺-release channel. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1992-2005. [PMID: 25499268 DOI: 10.1016/j.bbamcr.2014.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 12/19/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) type 2 (IP3R2) is an intracellular Ca²⁺-release channel located on the endoplasmic reticulum (ER). IP3R2 is characterized by a high sensitivity to both IP3 and ATP and is biphasically regulated by Ca²⁺. Furthermore, IP3R2 is modulated by various protein kinases. In addition to its regulation by protein kinase A, IP3R2 forms a complex with adenylate cyclase 6 and is directly regulated by cAMP. Finally, in the ER, IP3R2 is less mobile than the other IP3R isoforms, while its functional properties appear dominant in heterotetramers. These properties make the IP3R2 a Ca²⁺ channel with exquisite properties for setting up intracellular Ca²⁺ signals with unique characteristics. IP3R2 plays a crucial role in the function of secretory cell types (e.g. pancreatic acinar cells, hepatocytes, salivary gland, eccrine sweat gland). In cardiac myocytes, the role of IP3R2 appears more complex, because, together with IP3R1, it is needed for normal cardiogenesis, while its aberrant activity is implicated in cardiac hypertrophy and arrhythmias. Most importantly, its high sensitivity to IP3 makes IP3R2 a target for anti-apoptotic proteins (e.g. Bcl-2) in B-cell cancers. Disrupting IP3R/Bcl-2 interaction therefore leads in those cells to increased Ca²⁺ release and apoptosis. Intriguingly, IP3R2 is not only implicated in apoptosis but also in the induction of senescence, another tumour-suppressive mechanism. These results were the first to unravel the physiological and pathophysiological role of IP3R2 and we anticipate that further progress will soon be made in understanding the function of IP3R2 in various tissues and organs.
Collapse
Affiliation(s)
- Tamara Vervloessem
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - David I Yule
- University of Rochester, Department of Pharmacology and Physiology, Rochester, NY, USA
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium.
| |
Collapse
|
25
|
Mikoshiba K. Role of IP3 receptor signaling in cell functions and diseases. Adv Biol Regul 2014; 57:217-27. [PMID: 25497594 DOI: 10.1016/j.jbior.2014.10.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 10/06/2014] [Indexed: 11/25/2022]
Abstract
IP3 receptor (IP3R) was found to release Ca(2+) from non-mitochondrial store but the exact localization and the mode of action of IP3 remained a mystery. IP3R was identified to be P400 protein, a protein, which was missing in the cerebellum of ataxic mutant mice lacking Ca(2+) spikes in Pukinje cells. IP3R was an IP3 binding protein and was a Ca(2+) channel localized on the endoplasmic reticulum. Full-length cDNA of IP3R type 1 was initially cloned and later two other isoforms of IP3R (IP3R type 2 and type 3) were cloned in vertebrates. Interestingly, the phosphorylation sites, splicing sites, associated molecules, IP3 binding affinity and 5' promoter sequences of each isoform were different. Thus each isoform of IP3 receptor plays a role as a signaling hub offering a unique platform for matching various functional molecules that determines different trajectories of cell signaling. Because of this distinct role of each isoform of IP3R, the dysregulation of IP3 receptor causes various kinds of diseases in human and rodents such as ataxia, vulnerability to neuronal degeneration, heart disease, exocrine secretion deficit, taste perception deficit. Moreover, IP3 was found not only to release Ca(2+), but also to release IRBIT (IP3receptor binding protein released with inositol trisphosphate) essential for the regulation of acid-base balance, RNA synthesis and ribonucleotide reductase.
Collapse
Affiliation(s)
- Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, Japan.
| |
Collapse
|
26
|
Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev 2012; 92:537-76. [PMID: 22535891 DOI: 10.1152/physrev.00027.2011] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protein folding is a complex, error-prone process that often results in an irreparable protein by-product. These by-products can be recognized by cellular quality control machineries and targeted for proteasome-dependent degradation. The folding of proteins in the secretory pathway adds another layer to the protein folding "problem," as the endoplasmic reticulum maintains a unique chemical environment within the cell. In fact, a growing number of diseases are attributed to defects in secretory protein folding, and many of these by-products are targeted for a process known as endoplasmic reticulum-associated degradation (ERAD). Since its discovery, research on the mechanisms underlying the ERAD pathway has provided new insights into how ERAD contributes to human health during both normal and diseases states. Links between ERAD and disease are evidenced from the loss of protein function as a result of degradation, chronic cellular stress when ERAD fails to keep up with misfolded protein production, and the ability of some pathogens to coopt the ERAD pathway. The growing number of ERAD substrates has also illuminated the differences in the machineries used to recognize and degrade a vast array of potential clients for this pathway. Despite all that is known about ERAD, many questions remain, and new paradigms will likely emerge. Clearly, the key to successful disease treatment lies within defining the molecular details of the ERAD pathway and in understanding how this conserved pathway selects and degrades an innumerable cast of substrates.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
27
|
Takagaki Y, Yamagishi H, Matsuoka R. Factors Involved in Signal Transduction During Vertebrate Myogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:187-272. [DOI: 10.1016/b978-0-12-394307-1.00004-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
The Discovery and Structural Investigation of the IP3 Receptor and the Associated IRBIT Protein. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:281-304. [DOI: 10.1007/978-94-007-2888-2_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
29
|
Kodo K, Yamagishi H. A decade of advances in the molecular embryology and genetics underlying congenital heart defects. Circ J 2011; 75:2296-304. [PMID: 21914956 DOI: 10.1253/circj.cj-11-0636] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital heart defects (CHD) are the most common type of human birth defect and result in significant mortality worldwide. Despite numerous epidemiologic studies in the past decades, few genetic causes have been identified until recently. CHD result from abnormal morphogenesis of the systematic cardiovascular construction during development. Recent advances in molecular embryology, including the discovery of a new source of cardiac progenitor cells termed the second heart field (SHF), have revealed that the heart arises from multiple distinct embryonic origins. Cells derived from the SHF contribute to the development of the cardiac outflow tract, together with the other progenitor cell lineage called cardiac neural crest cells. Numerous cardiac transcription factors regulate these progenitor cells during heart development. Elucidation of the transcriptional network for these cardiac progenitor cells is essential for further understanding cardiac development and providing new insights into the morphogenesis of CHD. This review outlines the recent discoveries of the molecular embryology of the normal heart and the genetic basis of CHD.
Collapse
Affiliation(s)
- Kazuki Kodo
- Department of Pediatrics, Division of Pediatric Cardiology, Keio University School of Medicine, Japan
| | | |
Collapse
|