1
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Zhang B, Zhu Y, Zhang Z, Wu F, Ma X, Sheng W, Dai R, Guo Z, Yan W, Hao L, Huang G, Ma D, Hao B, Ma J. SMC3 contributes to heart development by regulating super-enhancer associated genes. Exp Mol Med 2024; 56:1826-1842. [PMID: 39085358 PMCID: PMC11372143 DOI: 10.1038/s12276-024-01293-0] [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: 09/29/2023] [Revised: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 08/02/2024] Open
Abstract
Abnormal cardiac development has been observed in individuals with Cornelia de Lange syndrome (CdLS) due to mutations in genes encoding members of the cohesin complex. However, the precise role of cohesin in heart development remains elusive. In this study, we aimed to elucidate the indispensable role of SMC3, a component of the cohesin complex, in cardiac development and its underlying mechanism. Our investigation revealed that CdLS patients with SMC3 mutations have high rates of congenital heart disease (CHD). We utilized heart-specific Smc3-knockout (SMC3-cKO) mice, which exhibit varying degrees of outflow tract (OFT) abnormalities, to further explore this relationship. Additionally, we identified 16 rare SMC3 variants with potential pathogenicity in individuals with isolated CHD. By employing single-nucleus RNA sequencing and chromosome conformation capture high-throughput genome-wide translocation sequencing, we revealed that Smc3 deletion downregulates the expression of key genes, including Ets2, in OFT cardiac muscle cells by specifically decreasing interactions between super-enhancers (SEs) and promoters. Notably, Ets2-SE-null mice also exhibit delayed OFT development in the heart. Our research revealed a novel role for SMC3 in heart development via the regulation of SE-associated genes, suggesting its potential relevance as a CHD-related gene and providing crucial insights into the molecular basis of cardiac development.
Collapse
Affiliation(s)
- Bowen Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Yongchang Zhu
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Feizhen Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Xiaojing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Wei Sheng
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Ranran Dai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Zhenglong Guo
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Weili Yan
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Lili Hao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Guoying Huang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Bingtao Hao
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Henan Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450000, China.
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Sanches BDA, Teófilo FBS, Brunet MY, Villapun VM, Man K, Rocha LC, Neto JP, Matsumoto MR, Maldarine JS, Ciena AP, Cox SC, Carvalho HF. Telocytes: current methods of research, challenges and future perspectives. Cell Tissue Res 2024; 396:141-155. [PMID: 38539007 DOI: 10.1007/s00441-024-03888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/12/2024] [Indexed: 04/28/2024]
Abstract
Telocytes (TCs) are CD34-positive interstitial cells that have long cytoplasmic projections, called telopodes; they have been identified in several organs and in various species. These cells establish a complex communication network between different stromal and epithelial cell types, and there is growing evidence that they play a key role in physiology and pathology. In many tissues, TC network impairment has been implicated in the onset and progression of pathological conditions, which makes the study of TCs of great interest for the development of novel therapies. In this review, we summarise the main methods involved in the characterisation of these cells as well as their inherent difficulties and then discuss the functional assays that are used to uncover the role of TCs in normal and pathological conditions, from the most traditional to the most recent. Furthermore, we provide future perspectives in the study of TCs, especially regarding the establishment of more precise markers, commercial lineages and means for drug delivery and genetic editing that directly target TCs.
Collapse
Affiliation(s)
- Bruno D A Sanches
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Francisco B S Teófilo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Victor M Villapun
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Lara C Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Marta R Matsumoto
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Juliana S Maldarine
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Adriano P Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil.
| |
Collapse
|
4
|
Mehta Z, Touma M. Post-Transcriptional Modification by Alternative Splicing and Pathogenic Splicing Variants in Cardiovascular Development and Congenital Heart Defects. Int J Mol Sci 2023; 24:ijms24021555. [PMID: 36675070 PMCID: PMC9862068 DOI: 10.3390/ijms24021555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Advancements in genomics, bioinformatics, and genome editing have uncovered new dimensions in gene regulation. Post-transcriptional modifications by the alternative splicing of mRNA transcripts are critical regulatory mechanisms of mammalian gene expression. In the heart, there is an expanding interest in elucidating the role of alternative splicing in transcriptome regulation. Substantial efforts were directed toward investigating this process in heart development and failure. However, few studies shed light on alternative splicing products and their dysregulation in congenital heart defects (CHDs). While elegant reports showed the crucial roles of RNA binding proteins (RBPs) in orchestrating splicing transitions during heart development and failure, the impact of RBPs dysregulation or genetic variation on CHDs has not been fully addressed. Herein, we review the current understanding of alternative splicing and RBPs' roles in heart development and CHDs. Wediscuss the impact of perinatal splicing transition and its dysregulation in CHDs. We further summarize the discoveries made of causal splicing variants in key transcription factors that are implicated in CHDs. An improved understanding of the roles of alternative splicing in heart development and CHDs may potentially inform novel preventive and therapeutic advancements for newborn infants with CHDs.
Collapse
Affiliation(s)
- Zubin Mehta
- Neonatal/Congenital Heart Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Children’s Discovery and Innovation Institute, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Marlin Touma
- Neonatal/Congenital Heart Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Children’s Discovery and Innovation Institute, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence:
| |
Collapse
|
5
|
Halmetoja E, Nagy I, Szabo Z, Alakoski T, Yrjölä R, Vainio L, Viitavaara E, Lin R, Rahtu-Korpela L, Vainio S, Kerkelä R, Magga J. Wnt11 in regulation of physiological and pathological cardiac growth. FASEB J 2022; 36:e22544. [PMID: 36098469 DOI: 10.1096/fj.202101856rrrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/11/2022]
Abstract
Wnt11 regulates early cardiac development and left ventricular compaction in the heart, but it is not known how Wnt11 regulates postnatal cardiac maturation and response to cardiac stress in the adult heart. We studied cell proliferation/maturation in postnatal and adolescent Wnt11 deficient (Wnt11-/-) heart and subjected adult mice with partial (Wnt11+/-) and complete Wnt11 (Wnt11-/-) deficiency to cardiac pressure overload. In addition, we subjected primary cardiomyocytes to recombinant Wnt proteins to study their effect on cardiomyocyte growth. Wnt11 deficiency did not affect cardiomyocyte proliferation or maturation in the postnatal or adolescent heart. However, Wnt11 deficiency led to enlarged heart phenotype that was not accompanied by significant hypertrophy of individual cardiomyocytes. Analysis of stressed adult hearts from wild-type mice showed a progressive decrease in Wnt11 expression in response to pressure overload. When studied in experimental cardiac pressure overload, Wnt11 deficiency did not exacerbate cardiac hypertrophy or remodeling and cardiac function remained identical between the genotypes. When subjecting cardiomyocytes to hypertrophic stimulus, the presence of recombinant Wnt11 together with Wnt5a reduced protein synthesis. In conclusion, Wnt11 deficiency does not affect postnatal cardiomyocyte proliferation but leads to cardiac growth. Interestingly, Wnt11 deficiency alone does not substantially modulate hypertrophic response to pressure overload in vivo. Wnt11 may require cooperation with other noncanonical Wnt proteins to regulate hypertrophic response under stress.
Collapse
Affiliation(s)
| | - Irina Nagy
- Department of Clinical Chemistry, Cancer and Translational Medicine Research Unit, Medical Research Center, University of Oulu and Northern Finland Laboratory Centre NordLab, Oulu University Hospital, Oulu, Finland
| | - Zoltan Szabo
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Tarja Alakoski
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Raisa Yrjölä
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Laura Vainio
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | | | - Ruizhu Lin
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | | | - Seppo Vainio
- Laboratory of Developmental Biology, Center for Cell Matrix Research, University of Oulu, Oulu, Finland.,Kvantum Institute, Infotech Oulu, University of Oulu, Oulu, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Johanna Magga
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Cardiovascular diseases are the leading cause of death worldwide, largely due to the limited regenerative capacity of the adult human heart. In contrast, teleost zebrafish hearts possess natural regeneration capacity by proliferation of pre-existing cardiomyocytes after injury. Hearts of mice can regenerate if injured in a few days after birth, which coincides with the transient capacity for cardiomyocyte proliferation. This review tends to elaborate the roles and mechanisms of Wnt/β-catenin signaling in heart development and regeneration in mammals and non-mammalian vertebrates. RECENT FINDINGS Studies in zebrafish, mice, and human embryonic stem cells demonstrate the binary effect for Wnt/β-catenin signaling during heart development. Both Wnts and Wnt antagonists are induced in multiple cell types during cardiac development and injury repair. In this review, we summarize composites of the Wnt signaling pathway and their different action routes, followed by the discussion of their involvements in cardiac specification, proliferation, and patterning. We provide overviews about canonical and non-canonical Wnt activity during heart homeostasis, remodeling, and regeneration. Wnt/β-catenin signaling exhibits biphasic and antagonistic effects on cardiac specification and differentiation depending on the stage of embryogenesis. Inhibition of Wnt signaling is beneficial for cardiac wound healing and functional recovery after injury. Understanding of the roles and mechanisms of Wnt signaling pathway in injured animal hearts will contribute to the development of potential therapeutics for human diseased hearts.
Collapse
Affiliation(s)
- Dongliang Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jianjian Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China
| | - Tao P Zhong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
7
|
Cucu I, Nicolescu MI, Busnatu ȘS, Manole CG. Dynamic Involvement of Telocytes in Modulating Multiple Signaling Pathways in Cardiac Cytoarchitecture. Int J Mol Sci 2022; 23:5769. [PMID: 35628576 PMCID: PMC9143034 DOI: 10.3390/ijms23105769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
Cardiac interstitium is a complex and dynamic environment, vital for normal cardiac structure and function. Telocytes are active cellular players in regulating main events that feature myocardial homeostasis and orchestrating its involvement in heart pathology. Despite the great amount of data suggesting (microscopically, proteomically, genetically, etc.) the implications of telocytes in the different physiological and reparatory/regenerative processes of the heart, understanding their involvement in realizing the heart's mature cytoarchitecture is still at its dawn. Our scrutiny of the recent literature gave clearer insights into the implications of telocytes in the WNT signaling pathway, but also TGFB and PI3K/AKT pathways that, inter alia, conduct cardiomyocytes differentiation, maturation and final integration into heart adult architecture. These data also strengthen evidence for telocytes as promising candidates for cellular therapies in various heart pathologies.
Collapse
Affiliation(s)
- Ioana Cucu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihnea Ioan Nicolescu
- Division of Histology, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Laboratory of Radiobiology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Ștefan-Sebastian Busnatu
- Department of Cardiology-“Bagdasar Arseni” Emergency Clinical Hospital, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 041915 Bucharest, Romania
| | - Cătălin Gabriel Manole
- Department of Cellular & Molecular Biology and Histology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Laboratory of Ultrastructural Pathology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
8
|
Caetano da Silva C, Edouard T, Fradin M, Aubert-Mucca M, Ricquebourg M, Raman R, Salles JP, Charon V, Guggenbuhl P, Muller M, Cohen-Solal M, Collet C. WNT11, a new gene associated with early onset osteoporosis, is required for osteoblastogenesis. Hum Mol Genet 2022; 31:1622-1634. [PMID: 34875064 PMCID: PMC9122655 DOI: 10.1093/hmg/ddab349] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Monogenic early onset osteoporosis (EOOP) is a rare disease defined by low bone mineral density (BMD) that results in increased risk of fracture in children and young adults. Although several causative genes have been identified, some of the EOOP causation remains unresolved. Whole-exome sequencing revealed a de novo heterozygous loss-of-function mutation in Wnt family member 11 (WNT11) (NM_004626.2:c.677_678dup p.Leu227Glyfs*22) in a 4-year-old boy with low BMD and fractures. We identified two heterozygous WNT11 missense variants (NM_004626.2:c.217G > A p.Ala73Thr) and (NM_004626.2:c.865G > A p.Val289Met) in a 51-year-old woman and in a 61-year-old woman, respectively, both with bone fragility. U2OS cells with heterozygous WNT11 mutation (NM_004626.2:c.690_721delfs*40) generated by CRISPR-Cas9 showed reduced cell proliferation (30%) and osteoblast differentiation (80%) as compared with wild-type U2OS cells. The expression of genes in the Wnt canonical and non-canonical pathways was inhibited in these mutant cells, but recombinant WNT11 treatment rescued the expression of Wnt pathway target genes. Furthermore, the expression of RSPO2, a WNT11 target involved in bone cell differentiation, and its receptor leucine-rich repeat containing G protein-coupled receptor 5 (LGR5), was decreased in WNT11 mutant cells. Treatment with WNT5A and WNT11 recombinant proteins reversed LGR5 expression, but Wnt family member 3A (WNT3A) recombinant protein treatment had no effect on LGR5 expression in mutant cells. Moreover, treatment with recombinant RSPO2 but not WNT11 or WNT3A activated the canonical pathway in mutant cells. In conclusion, we have identified WNT11 as a new gene responsible for EOOP, with loss-of-function variant inhibiting bone formation via Wnt canonical and non-canonical pathways. WNT11 may activate Wnt signaling by inducing the RSPO2-LGR5 complex via the non-canonical Wnt pathway.
Collapse
Affiliation(s)
- Caroline Caetano da Silva
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Thomas Edouard
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Melanie Fradin
- Service de Génétique Clinique, Centre de Référence des Anomalies du Développement de l'Ouest, Hôpital Sud de Rennes, Rennes F-35033, France
| | - Marion Aubert-Mucca
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Manon Ricquebourg
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Ratish Raman
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège 4000, Belgium
| | - Jean Pierre Salles
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Valérie Charon
- Department of Radiology, CHU de Rennes, Rennes F-35000, France
| | | | - Marc Muller
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège 4000, Belgium
| | - Martine Cohen-Solal
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Corinne Collet
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
- Département de Génétique, UF de Génétique Moléculaire, Hôpital Robert Debré, APHP, Paris F-75019, France
| |
Collapse
|
9
|
Heilig AK, Nakamura R, Shimada A, Hashimoto Y, Nakamura Y, Wittbrodt J, Takeda H, Kawanishi T. Wnt11 acts on dermomyotome cells to guide epaxial myotome morphogenesis. eLife 2022; 11:71845. [PMID: 35522214 PMCID: PMC9075960 DOI: 10.7554/elife.71845] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 04/19/2022] [Indexed: 12/30/2022] Open
Abstract
The dorsal axial muscles, or epaxial muscles, are a fundamental structure covering the spinal cord and vertebrae, as well as mobilizing the vertebrate trunk. To date, mechanisms underlying the morphogenetic process shaping the epaxial myotome are largely unknown. To address this, we used the medaka zic1/zic4-enhancer mutant Double anal fin (Da), which exhibits ventralized dorsal trunk structures resulting in impaired epaxial myotome morphology and incomplete coverage over the neural tube. In wild type, dorsal dermomyotome (DM) cells reduce their proliferative activity after somitogenesis. Subsequently, a subset of DM cells, which does not differentiate into the myotome population, begins to form unique large protrusions extending dorsally to guide the epaxial myotome dorsally. In Da, by contrast, DM cells maintain the high proliferative activity and mainly form small protrusions. By combining RNA- and ChIP-sequencing analyses, we revealed direct targets of Zic1, which are specifically expressed in dorsal somites and involved in various aspects of development, such as cell migration, extracellular matrix organization, and cell-cell communication. Among these, we identified wnt11 as a crucial factor regulating both cell proliferation and protrusive activity of DM cells. We propose that dorsal extension of the epaxial myotome is guided by a non-myogenic subpopulation of DM cells and that wnt11 empowers the DM cells to drive the coverage of the neural tube by the epaxial myotome.
Collapse
Affiliation(s)
- Ann Kathrin Heilig
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan.,Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany.,Heidelberg Biosciences International Graduate School, Heidelberg, Germany
| | - Ryohei Nakamura
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Atsuko Shimada
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Yuka Hashimoto
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Yuta Nakamura
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Joachim Wittbrodt
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Toru Kawanishi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Guo R, Xing QS. Roles of Wnt Signaling Pathway and ROR2 Receptor in Embryonic Development: An Update Review Article. Epigenet Insights 2022; 15:25168657211064232. [PMID: 35128307 PMCID: PMC8808015 DOI: 10.1177/25168657211064232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022] Open
Abstract
The Wnt family is a large class of highly conserved cysteine-rich secretory glycoproteins that play a vital role in various cellular and physiological courses through different signaling pathways during embryogenesis and tissue homeostasis 3. Wnt5a is a secreted glycoprotein that belongs to the noncanonical Wnt family and is involved in a wide range of developmental and tissue homeostasis. A growing body of evidence suggests that Wnt5a affects embryonic development, signaling through various receptors, starting with the activation of β-catenin by Wnt5a. In addition to affecting planar cell polarity and Ca2+ pathways, β-catenin also includes multiple signaling cascades that regulate various cell functions. Secondly, Wnt5a can bind to Ror receptors to mediate noncanonical Wnt signaling and a significant ligand for Ror2 in vertebrates. Consistent with the multiple functions of Wnt5A/Ror2 signaling, Wnt5A knockout mice exhibited various phenotypic defects, including an inability to extend the anterior and posterior axes of the embryo. Numerous essential roles of Wnt5a/Ror2 in development have been demonstrated. Therefore, Ror signaling pathway become a necessary target for diagnosing and treating human diseases. The Wnt5a- Ror2 signaling pathway as a critical factor has attracted extensive attention.
Collapse
Affiliation(s)
- Rui Guo
- Qingdao University, Qingdao, China
| | - Quan Sheng Xing
- Qingdao University-Affiliated Hospital of Women and Children, Qingdao, China
- Quan Sheng Xing, Qingdao University-Affiliated Hospital of Women and Children, tongfu road 6, shibei district, Qingdao 266000, China.
| |
Collapse
|
11
|
Ma H, Liu Z, Yang Y, Feng D, Dong Y, Garbutt TA, Hu Z, Wang L, Luan C, Cooper CD, Li Y, Welch JD, Qian L, Liu J. Functional coordination of non-myocytes plays a key role in adult zebrafish heart regeneration. EMBO Rep 2021; 22:e52901. [PMID: 34523214 DOI: 10.15252/embr.202152901] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac regeneration occurs primarily through proliferation of existing cardiomyocytes, but also involves complex interactions between distinct cardiac cell types including non-cardiomyocytes (non-CMs). However, the subpopulations, distinguishing molecular features, cellular functions, and intercellular interactions of non-CMs in heart regeneration remain largely unexplored. Using the LIGER algorithm, we assemble an atlas of cell states from 61,977 individual non-CM scRNA-seq profiles isolated at multiple time points during regeneration. This analysis reveals extensive non-CM cell diversity, including multiple macrophage (MC), fibroblast (FB), and endothelial cell (EC) subpopulations with unique spatiotemporal distributions, and suggests an important role for MC in inducing the activated FB and EC subpopulations. Indeed, pharmacological perturbation of MC function compromises the induction of the unique FB and EC subpopulations. Furthermore, we developed computational algorithm Topologizer to map the topological relationships and dynamic transitions between functional states. We uncover dynamic transitions between MC functional states and identify factors involved in mRNA processing and transcriptional regulation associated with the transition. Together, our single-cell transcriptomic analysis of non-CMs during cardiac regeneration provides a blueprint for interrogating the molecular and cellular basis of this process.
Collapse
Affiliation(s)
- Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ziqing Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yuchen Yang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dong Feng
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanhan Dong
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tiffany A Garbutt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Li Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Changfei Luan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Cynthia D Cooper
- School of Molecular Biosciences, Washington State University Vancouver, Vancouver, WA, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA.,Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Zou Y, Pan L, Shen Y, Wang X, Huang C, Wang H, Jin X, Yin C, Wang Y, Jia J, Qian J, Zou Y, Gong H, Ge J. Cardiac Wnt5a and Wnt11 promote fibrosis by the crosstalk of FZD5 and EGFR signaling under pressure overload. Cell Death Dis 2021; 12:877. [PMID: 34564708 PMCID: PMC8464604 DOI: 10.1038/s41419-021-04152-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022]
Abstract
Progressive cardiac fibrosis accelerates the development of heart failure. Here, we aimed to explore serum Wnt5a and Wnt11 levels in hypertension patients, the roles of Wnt5a and Wnt11 in cardiac fibrosis and potential mechanisms under pressure overload. The pressure overload mouse model was built by transverse aortic constriction (TAC). Cardiac fibrosis was analyzed by Masson's staining. Serum Wnt5a or Wnt11 was elevated and associated with diastolic dysfunction in hypertension patients. TAC enhanced the expression and secretion of Wnt5a or Wnt11 from cardiomyocytes (CMs), cardiac fibroblasts (CFs), and cardiac microvascular endothelial cells (CMECs). Knockdown of Wnt5a and Wnt11 greatly improved cardiac fibrosis and function at 4 weeks after TAC. In vitro, shWnt5a or shWnt11 lentivirus transfection inhibited pro-fibrotic effects in CFs under mechanical stretch (MS). Similarly, conditional medium from stretched-CMs transfected with shWnt5a or shWnt11 lentivirus significantly suppressed the pro-fibrotic effects induced by conditional medium from stretched-CMs. These data suggested that CMs- or CFs-derived Wnt5a or Wnt11 showed a pro-fibrotic effect under pressure overload. In vitro, exogenous Wnt5a or Wnt11 activated ERK and p38 (fibrotic-related signaling) pathway, promoted the phosphorylation of EGFR, and increased the expression of Frizzled 5 (FZD5) in CFs. Inhibition or knockdown of EGFR greatly attenuated the increased FZD5, p-p38, and p-ERK levels, and the pro-fibrotic effect induced by Wnt5a or Wnt11 in CFs. Si-FZD5 transfection suppressed the increased p-EGFR level, and the fibrotic-related effects in CFs treated with Wnt5a or Wnt11. In conclusion, pressure overload enhances the secretion of Wnt5a or Wnt11 from CMs and CFs which promotes cardiac fibrosis by activation the crosstalk of FZD5 and EGFR. Thus, Wnt5a or Wnt11 may be a novel therapeutic target for the prevention of cardiac fibrosis under pressure overload.
Collapse
Affiliation(s)
- Yan Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Le Pan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yi Shen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenxing Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hao Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xuejuan Jin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianguo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Zhao Y, Wang LK, Eskin A, Kang X, Fajardo VM, Mehta Z, Pineles S, Schmidt RJ, Nagiel A, Satou G, Garg M, Federman M, Reardon LC, Lee SL, Biniwale R, Grody WW, Halnon N, Khanlou N, Quintero-Rivera F, Alejos JC, Nakano A, Fishbein GA, Van Arsdell GS, Nelson SF, Touma M. Recessive ciliopathy mutations in primary endocardial fibroelastosis: a rare neonatal cardiomyopathy in a case of Alstrom syndrome. J Mol Med (Berl) 2021; 99:1623-1638. [PMID: 34387706 PMCID: PMC8541947 DOI: 10.1007/s00109-021-02112-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022]
Abstract
Abstract Among neonatal cardiomyopathies, primary endocardial fibroelastosis (pEFE) remains a mysterious disease of the endomyocardium that is poorly genetically characterized, affecting 1/5000 live births and accounting for 25% of the entire pediatric dilated cardiomyopathy (DCM) with a devastating course and grave prognosis. To investigate the potential genetic contribution to pEFE, we performed integrative genomic analysis, using whole exome sequencing (WES) and RNA-seq in a female infant with confirmed pathological diagnosis of pEFE. Within regions of homozygosity in the proband genome, WES analysis revealed novel parent-transmitted homozygous mutations affecting three genes with known roles in cilia assembly or function. Among them, a novel homozygous variant [c.1943delA] of uncertain significance in ALMS1 was prioritized for functional genomic and mechanistic analysis. Loss of function mutations of ALMS1 have been implicated in Alstrom syndrome (AS) [OMIM 203800], a rare recessive ciliopathy that has been associated with cardiomyopathy. The variant of interest results in a frameshift introducing a premature stop codon. RNA-seq of the proband’s dermal fibroblasts confirmed the impact of the novel ALMS1 variant on RNA-seq reads and revealed dysregulated cellular signaling and function, including the induction of epithelial mesenchymal transition (EMT) and activation of TGFβ signaling. ALMS1 loss enhanced cellular migration in patient fibroblasts as well as neonatal cardiac fibroblasts, while ALMS1-depleted cardiomyocytes exhibited enhanced proliferation activity. Herein, we present the unique pathological features of pEFE compared to DCM and utilize integrated genomic analysis to elucidate the molecular impact of a novel mutation in ALMS1 gene in an AS case. Our report provides insights into pEFE etiology and suggests, for the first time to our knowledge, ciliopathy as a potential underlying mechanism for this poorly understood and incurable form of neonatal cardiomyopathy. Key message Primary endocardial fibroelastosis (pEFE) is a rare form of neonatal cardiomyopathy that occurs in 1/5000 live births with significant consequences but unknown etiology. Integrated genomics analysis (whole exome sequencing and RNA sequencing) elucidates novel genetic contribution to pEFE etiology. In this case, the cardiac manifestation in Alstrom syndrome is pEFE. To our knowledge, this report provides the first evidence linking ciliopathy to pEFE etiology. Infants with pEFE should be examined for syndromic features of Alstrom syndrome. Our findings lead to a better understanding of the molecular mechanisms of pEFE, paving the way to potential diagnostic and therapeutic applications.
Supplementary information The online version contains supplementary material available at 10.1007/s00109-021-02112-z.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lee-Kai Wang
- Institute for Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ascia Eskin
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xuedong Kang
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Viviana M Fajardo
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Zubin Mehta
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stacy Pineles
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan J Schmidt
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Gary Satou
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Meena Garg
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Myke Federman
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Leigh C Reardon
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Ahmanson/UCLA Adult Congenital Heart Disease Center, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Steven L Lee
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Reshma Biniwale
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Wayne W Grody
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nancy Halnon
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Negar Khanlou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine and Department of Pediatrics, University of California Irvine, CA, Irvine, USA
| | - Juan C Alejos
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Atsushi Nakano
- Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gregory A Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Glen S Van Arsdell
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Institute for Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marlin Touma
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA. .,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,The Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Prashanth G, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules. BMC Endocr Disord 2021; 21:80. [PMID: 33902539 PMCID: PMC8074411 DOI: 10.1186/s12902-021-00718-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. METHODS To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. RESULTS A total of 820 DEGs were identified between healthy obese and metabolically unhealthy obese, among 409 up regulated and 411 down regulated genes. The GO enrichment analysis results showed that these DEGs were significantly enriched in ion transmembrane transport, intrinsic component of plasma membrane, transferase activity, transferring phosphorus-containing groups, cell adhesion, integral component of plasma membrane and signaling receptor binding, whereas, the REACTOME pathway enrichment analysis results showed that these DEGs were significantly enriched in integration of energy metabolism and extracellular matrix organization. The hub genes CEBPD, TP73, ESR2, TAB1, MAP 3K5, FN1, UBD, RUNX1, PIK3R2 and TNF, which might play an essential role in obesity associated type 2 diabetes mellitus was further screened. CONCLUSIONS The present study could deepen the understanding of the molecular mechanism of obesity associated type 2 diabetes mellitus, which could be useful in developing therapeutic targets for obesity associated type 2 diabetes mellitus.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, Karnataka, 577501, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka, 582209, India
| |
Collapse
|
15
|
Zhao Y, Kang X, Barsegian A, He J, Guzman A, Lau RP, Biniwale R, Wadhra M, Reemtsen B, Garg M, Halnon N, Quintero-Rivera F, Grody WW, Van Arsdell G, Nelson SF, Touma M. Gene-environment regulation of chamber-specific maturation during hypoxemic perinatal circulatory transition. J Mol Med (Berl) 2020; 98:1009-1020. [PMID: 32533200 DOI: 10.1007/s00109-020-01933-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Chamber-specific and temporally regulated perinatal cardiac growth and maturation is critical for functional adaptation of the heart and may be altered significantly in response to perinatal stress, such as systemic hypoxia (hypoxemia), leading to significant pathology, even mortality. Understanding transcriptome regulation of neonatal heart chambers in response to hypoxemia is necessary to develop chamber-specific therapies for infants with cyanotic congenital heart defects (CHDs). We sought to determine chamber-specific transcriptome programming during hypoxemic perinatal circulatory transition. We performed transcriptome-wide analysis on right ventricle (RV) and left ventricle (LV) of postnatal day 3 (P3) mouse hearts exposed to perinatal hypoxemia. Hypoxemia decreased baseline differences between RV and LV leading to significant attenuation of ventricular patterning (AVP), which involved several molecular pathways, including Wnt signaling suppression and cell cycle induction. Notably, robust changes in RV transcriptome in hypoxemic condition contributed significantly to the AVP. Remarkably, suppression of epithelial mesenchymal transition (EMT) and dysregulation of the TP53 signaling were prominent hallmarks of the AVP genes in neonatal mouse heart. Furthermore, members of the TP53-related gene family were dysregulated in the hypoxemic RVs of neonatal mouse and cyanotic Tetralogy of Fallot hearts. Integrated analysis of chamber-specific transcriptome revealed hypoxemia-specific changes that were more robust in RVs compared with LVs, leading to previously uncharacterized AVP induced by perinatal hypoxemia. Remarkably, reprogramming of EMT process and dysregulation of the TP53 network contributed to transcriptome remodeling of neonatal heart during hypoxemic circulatory transition. These insights may enhance our understanding of hypoxemia-induced pathogenesis in newborn infants with cyanotic CHD phenotypes. KEY MESSAGES: During perinatal circulatory transition, transcriptome programming is a major driving force of cardiac chamber-specific maturation and adaptation to hemodynamic load and external environment. During hypoxemic perinatal transition, transcriptome reprogramming may affect chamber-specific growth and development, particularly in newborns with congenital heart defects (CHDs). Chamber-specific transcriptome changes during hypoxemic perinatal transition are yet to be fully elucidated. Systems-based analysis of hypoxemic neonatal hearts at postnatal day 3 reveals chamber-specific transcriptome signatures during hypoxemic perinatal transition, which involve attenuation of ventricular patterning (AVP) and repression of epithelial mesenchymal transition (EMT). Key regulatory circuits involved in hypoxemia response were identified including suppression of Wnt signaling, induction of cellular proliferation and dysregulation of TP53 network.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Xuedong Kang
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Alexander Barsegian
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Jian He
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Alejandra Guzman
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan P Lau
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Reshma Biniwale
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Madhuri Wadhra
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Brian Reemtsen
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Meena Garg
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA
| | - Nancy Halnon
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Wayne W Grody
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Glen Van Arsdell
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marlin Touma
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles 675 Charles E. Young Dr S, 3762 MacDonald Research Laboratories, Los Angeles, CA, 90024, USA. .,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Children's Discovery and Innovation Institute, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,The Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA. .,Eli and Edythe Stem Cell Institute, University of California Los Angeles, Los Angeles, CA, USA. .,Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Mohamed T, Abdul-Hafez A, Gewolb IH, Uhal BD. Oxygen injury in neonates: which is worse? hyperoxia, hypoxia, or alternating hyperoxia/hypoxia. JOURNAL OF LUNG, PULMONARY & RESPIRATORY RESEARCH 2020; 7:4-13. [PMID: 34337150 PMCID: PMC8320601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Premature birth results in an increased risk of respiratory distress and often requires oxygen therapy. While the supplemental oxygen has been implicated as a cause of bronchopulmonary dysplasia (BPD), in clinical practice this supplementation usually only occurs after the patient's oxygen saturation levels have dropped. The effect of hyperoxia on neonates has been extensively studied. However, there is an unanswered fundamental question: which has the most impact-hyperoxia, hypoxia or fluctuating oxygen levels? In this review, we will summarize the reported effect of hypoxia, hyperoxia or a fluctuation of oxygen levels (hypoxia/hyperoxia cycling) in preterm neonates, with special emphasis on the lungs.
Collapse
Affiliation(s)
- Tarek Mohamed
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Amal Abdul-Hafez
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Ira H Gewolb
- Department of Pediatrics and Human Development, Michigan State University, USA
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, USA
| |
Collapse
|
17
|
Mohamed IA, El-Badri N, Zaher A. Wnt Signaling: The double-edged sword diminishing the potential of stem cell therapy in congenital heart disease. Life Sci 2019; 239:116937. [PMID: 31629761 DOI: 10.1016/j.lfs.2019.116937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022]
Abstract
Stem cell therapy using bone marrow derived or mesenchymal stem cells has become a popular option for cardiovascular disease treatment, however the administration of embryonic stem cells has been mostly experimental. Remarkably, most of these ongoing clinical trials involve adult patients, but little is known regarding the safety and efficacy of stem cell therapy in newborns and children battling congenital heart diseases. Furthermore, cell delivery methods involve the administration of stem cells without pre-differentiation, and without consideration for the consequent process of cardiac development. Interestingly, in-vitro studies have demonstrated that the differentiation of embryonic stem cells into cardiomyocytes imitates the stages of cardiogenesis. Wnt signaling plays a profound role during the earliest stages of cardiogenesis and cardiac differentiation. In fact inappropriate Wnt signaling is associated with numerous cardiac disorders especially congenital heart disease. Furthermore, cell-extracellular matrix interactions were shown to be critical for stem cell differentiation and adequate cardiogenesis. Since extracellular matrix molecules are fundamental for maintenance and repair during heart disease and congenital heart disease, they may offer a novel approach for therapy. Herein we aim to review the critical role of Wnt signaling, as well as the profound importance of cell extracellular matrix interaction, during cardiogenesis. Both of these processes are crucial for precise stem cell differentiation into cardiomyocytes and developing efficacious regenerative therapy for congenital heart disease.
Collapse
Affiliation(s)
- Iman A Mohamed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, 12588, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, 12588, Egypt
| | - Amr Zaher
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 6th of October City, 12588, Egypt; National Heart Institute, Giza, Egypt.
| |
Collapse
|
18
|
Zhao Y, Kang X, Gao F, Guzman A, Lau RP, Biniwale R, Wadehra M, Reemtsen B, Garg M, Halnon N, Quintero-Rivera F, Van Arsdell G, Coppola G, Nelson SF, Touma M. Gene-environment regulatory circuits of right ventricular pathology in tetralogy of fallot. J Mol Med (Berl) 2019; 97:1711-1722. [PMID: 31834445 DOI: 10.1007/s00109-019-01857-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/02/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022]
Abstract
The phenotypic spectrum of congenital heart defects (CHDs) is contributed by both genetic and environmental factors. Their interactions are profoundly heterogeneous but may operate on common pathways as in the case of hypoxia signaling during postnatal heart development in the context of CHDs. Tetralogy of Fallot (TOF) is the most common cyanotic (hypoxemic) CHD. However, how the hypoxic environment contributes to TOF pathogenesis after birth is poorly understood. We performed Genome-wide transcriptome analysis on right ventricle outflow tract (RVOT) specimens from cyanotic and noncyanotic TOF. Co-expression network analysis identified gene modules specifically associated with clinical diagnosis and hypoxemia status in the TOF hearts. In particular, hypoxia-dependent induction of myocyte proliferation is associated with E2F1-mediated cell cycle regulation and repression of the WNT11-RB1 axis. Genes enriched in epithelial mesenchymal transition (EMT), fibrosis, and sarcomere were also repressed in cyanotic TOF patients. Importantly, transcription factor analysis of the hypoxia-regulated modules suggested CREB1 as a putative regulator of hypoxia/WNT11-RB1 circuit. The study provides a high-resolution landscape of transcriptome programming associated with TOF phenotypes and unveiled hypoxia-induced regulatory circuit in cyanotic TOF. Hypoxia-induced cardiomyocyte proliferation involves negative modulation of CREB1 activity upstream of the WNT11-RB1 axis. KEY MESSAGES: Genetic and environmental factors contribute to congenital heart defects (CHDs). How hypoxia contributes to Tetralogy of Fallot (TOF) pathogenesis after birth is unclear. Systems biology-based analysis revealed distinct molecular signature in CHDs. Gene expression modules specifically associated with cyanotic TOF were uncovered. Key regulatory circuits induced by hypoxia in TOF pathogenesis after birth were unveiled.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California, Los Angeles, CA, USA
| | - Xuedong Kang
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California, Los Angeles, CA, USA
| | - Fuying Gao
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Alejandra Guzman
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA
| | - Ryan P Lau
- Department of Pathology and Laboratory Medicine, Clinical Genomics Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Reshma Biniwale
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Madhuri Wadehra
- Department of Pathology and Laboratory Medicine, Clinical Genomics Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Brian Reemtsen
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Meena Garg
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA
| | - Nancy Halnon
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine, Clinical Genomics Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Glen Van Arsdell
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Stanley F Nelson
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Human Genetics, Institute of Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Institute of Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Marlin Touma
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833 Le Conte Ave, MDCC-B2-375, Los Angeles, CA, 90095, USA. .,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California, Los Angeles, CA, USA. .,Department of Human Genetics, Institute of Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA. .,Institute of Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA. .,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA. .,The Molecular Biology Institute, University of California, Los Angeles, CA, USA. .,Eli and Edythe Stem Cell Institute, University of California, Los Angeles, CA, USA.
| | | |
Collapse
|
19
|
Bittle GJ, Morales D, Deatrick KB, Parchment N, Saha P, Mishra R, Sharma S, Pietris N, Vasilenko A, Bor C, Ambastha C, Gunasekaran M, Li D, Kaushal S. Stem Cell Therapy for Hypoplastic Left Heart Syndrome: Mechanism, Clinical Application, and Future Directions. Circ Res 2019; 123:288-300. [PMID: 29976693 DOI: 10.1161/circresaha.117.311206] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypoplastic left heart syndrome is a type of congenital heart disease characterized by underdevelopment of the left ventricle, outflow tract, and aorta. The condition is fatal if aggressive palliative operations are not undertaken, but even after the complete 3-staged surgical palliation, there is significant morbidity because of progressive and ultimately intractable right ventricular failure. For this reason, there is interest in developing novel therapies for the management of right ventricular dysfunction in patients with hypoplastic left heart syndrome. Stem cell therapy may represent one such innovative approach. The field has identified numerous stem cell populations from different tissues (cardiac or bone marrow or umbilical cord blood), different age groups (adult versus neonate-derived), and different donors (autologous versus allogeneic), with preclinical and clinical experience demonstrating the potential utility of each cell type. Preclinical trials in small and large animal models have elucidated several mechanisms by which stem cells affect the injured myocardium. Our current understanding of stem cell activity is undergoing a shift from a paradigm based on cellular engraftment and differentiation to one recognizing a primarily paracrine effect. Recent studies have comprehensively evaluated the individual components of the stem cells' secretomes, shedding new light on the intracellular and extracellular pathways at the center of their therapeutic effects. This research has laid the groundwork for clinical application, and there are now several trials of stem cell therapies in pediatric populations that will provide important insights into the value of this therapeutic strategy in the management of hypoplastic left heart syndrome and other forms of congenital heart disease. This article reviews the many stem cell types applied to congenital heart disease, their preclinical investigation and the mechanisms by which they might affect right ventricular dysfunction in patients with hypoplastic left heart syndrome, and finally, the completed and ongoing clinical trials of stem cell therapy in patients with congenital heart disease.
Collapse
Affiliation(s)
- Gregory J Bittle
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - David Morales
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Kristopher B Deatrick
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Nathaniel Parchment
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Progyaparamita Saha
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Rachana Mishra
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Sudhish Sharma
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Nicholas Pietris
- Division of Cardiology (N. Pietris), University of Maryland School of Medicine, Baltimore
| | - Alexander Vasilenko
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Casey Bor
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Chetan Ambastha
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Muthukumar Gunasekaran
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Deqiang Li
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Sunjay Kaushal
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| |
Collapse
|
20
|
Kerkhof PLM, Kuznetsova T, Yasha Kresh J, Handly N. Cardiophysiology Illustrated by Comparing Ventricular Volumes in Healthy Adult Males and Females. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:123-138. [PMID: 30051381 DOI: 10.1007/978-3-319-77932-4_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Recent advances in cardiac imaging techniques have substantially contributed to a growing interest in the analysis of global cardiac chamber dimensions and regional myocardial deformation. During the cardiac cycle, ventricular luminal volume varies due to the contraction process, which also confers a shape change including substantial alteration of long axis length, as well as rotation of the base compared to the apex. Local deformation can be assessed by strain (rate) analysis. Reviewing the present literature, it must be concluded that there is no single metric available to comprehensively characterize ventricular function. Every candidate advanced thus far has been found to incompletely reflect ventricular performance. This observation is not surprising in view of the complexity of the cardiac pump system. Additionally, sex-specific modifiers may play a role. More than three decades ago, it was shown that on average the ventricular volume is smaller in healthy women compared to matched males. Therefore, the present contribution concerns the interpretation of data derived from the healthy heart in both men and women. Starting from the classical Starling concept, we apply a simple mathematical transformation which permits an insightful representation of ventricular mechanics. Relating end-systolic volume (ESV) to end-diastolic volume creates the ventricular volume regulation graph which features the pertinent working point of an individual heart. This fundamental approach illustrates why certain proposed performance indexes cannot individually reveal the essence of ventricular systolic function. We demonstrate that particular metrics are highly interconnected and just tell us the same story in a different disguise. It is imperative to understand which associations exist and if they expectedly are (nearly) linear or frankly nonlinear. Notably, ejection fraction (EF) is primarily determined by ESV, while in turn EF is not much different from ventriculo-arterial coupling (VAC). Insight into cardiac function is promoted by identification of the paramount/essential components involved. The smaller ESV (p < 0.0001) implies that EF is higher in women and may also have consequences for VAC.
Collapse
Affiliation(s)
- Peter L M Kerkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands.
| | - Tatiana Kuznetsova
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - J Yasha Kresh
- Departments of Cardiothoracic Surgery and Medicine (Cardiology), Drexel University College of Medicine, IME, University of Pennsylvania, Philadelphia, PA, USA
| | - Neal Handly
- Department of Emergency Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|