1
|
Wang BX. Investigating Inherited Heart Diseases Using Human Induced Pluripotent Stem Cell-Based Models. Life (Basel) 2024; 14:1370. [PMID: 39598169 PMCID: PMC11595871 DOI: 10.3390/life14111370] [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: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Inherited heart diseases (IHDs) are caused by genetic mutations that disrupt the physiological structure and function of the heart. Understanding the mechanisms behind these diseases is crucial for developing personalised interventions in cardiovascular medicine. Development of induced pluripotent stem cells, which can then be differentiated to any nucleated adult cell type, has enabled the creation of personalised single-cell and multicellular models, providing unprecedented insights into the pathophysiology of IHDs. This review provides a comprehensive overview of recent advancements in human iPSC models used to dissect the molecular and genetic underpinnings of common IHDs. We examine multicellular models and tissue engineering approaches, such as cardiac organoids, engineered heart tissue, and multicellular co-culture systems, which simulate complex intercellular interactions within heart tissue. Recent advancements in stem cell models offer a more physiologically relevant platform to study disease mechanisms, enabling researchers to observe cellular interactions, study disease progression, and identify therapeutic strategies. By leveraging these innovative models, we can gain deeper insights into the molecular and cellular mechanisms underlying IHDs, ultimately paving the way for more effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Brian Xiangzhi Wang
- Department of Cardiology, Jersey General Hospital, Gloucester Street, St. Helier JE1 3QS, Jersey, UK
| |
Collapse
|
2
|
Zhou Y, Takahashi JI, Sakurai H. New Directions for Advanced Targeting Strategies of EGFR Signaling in Cancer. Biol Pharm Bull 2024; 47:895-903. [PMID: 38692865 DOI: 10.1248/bpb.b23-00924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Epidermal growth factor (EGF)-EGF receptor (EGFR) signaling studies paved the way for a basic understanding of growth factor and oncogene signaling pathways and the development of tyrosine kinase inhibitors (TKIs). Due to resistance mutations and the activation of alternative pathways when cancer cells escape TKIs, highly diverse cell populations form in recurrent tumors through mechanisms that have not yet been fully elucidated. In this review, we summarize recent advances in EGFR basic research on signaling networks and intracellular trafficking that may clarify the novel mechanisms of inhibitor resistance, discuss recent clinical developments in EGFR-targeted cancer therapy, and offer novel strategies for cancer drug development.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama
| | - Jun-Ichiro Takahashi
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama
| |
Collapse
|
3
|
Delogu AB, Limongelli G, Versacci P, Adorisio R, Kaski JP, Blandino R, Maiolo S, Monda E, Putotto C, De Rosa G, Chatfield KC, Gelb BD, Calcagni G. The heart in RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:440-451. [PMID: 36408797 DOI: 10.1002/ajmg.c.32014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/11/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022]
Abstract
The cardiovascular phenotype associated with RASopathies has expanded far beyond the original descriptions of pulmonary valve stenosis by Dr Jaqueline Noonan in 1968 and hypertrophic cardiomyopathy by Hirsch et al. in 1975. Because of the common underlying RAS/MAPK pathway dysregulation, RASopathy syndromes usually present with a typical spectrum of overlapping cardiovascular anomalies, although less common cardiac defects can occur. The identification of the causative genetic variants has enabled the recognition of specific correlations between genotype and cardiac phenotype. Characterization and understanding of genotype-phenotype associations is not only important for counseling a family of an infant with a new diagnosis of a RASopathy condition but is also critical for their clinical prognosis with respect to cardiac disease, neurodevelopment and other organ system involvement over the lifetime of the patient. This review will focus on the cardiac manifestations of the most common RASopathy syndromes, the relationship between cardiac defects and causal genetic variation, the contribution of cardiovascular abnormalities to morbidity and mortality and the most relevant follow-up issues for patients affected by RAS/MAPK pathway diseases, with respect to cardiac clinical outcomes and management, in children and in the adult population.
Collapse
Affiliation(s)
- Angelica Bibiana Delogu
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy.,European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paolo Versacci
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Rachele Adorisio
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Juan Pablo Kaski
- Centre for Pediatric Inherited and Rare Cardiovascular Disease, University College London Institute of Cardiovascular Science, London, UK.,Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London, UK
| | | | - Stella Maiolo
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy.,Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Emanuele Monda
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy.,European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Carolina Putotto
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Gabriella De Rosa
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Kathryn C Chatfield
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Departments of Pediatrics and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Giulio Calcagni
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| |
Collapse
|
4
|
Kontaridis MI, Chennappan S. Mitochondria and the future of RASopathies: the emergence of bioenergetics. J Clin Invest 2022; 132:1-5. [PMID: 35426371 PMCID: PMC9017150 DOI: 10.1172/jci157560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RASopathies are a family of rare autosomal dominant disorders that affect the canonical Ras/MAPK signaling pathway and manifest as neurodevelopmental systemic syndromes, including Costello syndrome (CS). In this issue of the JCI, Dard et al. describe the molecular determinants of CS using a myriad of genetically modified models, including mice expressing HRAS p.G12S, patient-derived skin fibroblasts, hiPSC-derived human cardiomyocytes, an HRAS p.G12V zebrafish model, and human lentivirally induced fibroblasts overexpressing HRAS p.G12S or HRAS p.G12A. Mitochondrial proteostasis and oxidative phosphorylation were altered in CS, and inhibition of the AMPK signaling pathway mediated bioenergetic changes. Importantly, the pharmacological induction of this pathway restored cardiac function and reduced the developmental defects associated with CS. These findings identify a role for altered bioenergetics and provide insights into more effective treatment strategies for patients with RASopathies.
Collapse
Affiliation(s)
- Maria I. Kontaridis
- Masonic Medical Research Institute, Department of Biological Sciences and Translational Medicine, Utica, New York, USA
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Cardiology, Boston, Massachusetts, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Saravanakkumar Chennappan
- Masonic Medical Research Institute, Department of Biological Sciences and Translational Medicine, Utica, New York, USA
| |
Collapse
|
5
|
Kong J, Long YQ. Recent advances in the discovery of protein tyrosine phosphatase SHP2 inhibitors. RSC Med Chem 2022; 13:246-257. [PMID: 35434626 PMCID: PMC8942255 DOI: 10.1039/d1md00386k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/14/2022] [Indexed: 01/17/2023] Open
Abstract
Src homology 2 domain-containing protein tyrosine phosphatase (SHP2) is a non-receptor protein tyrosine phosphatase encoded by the Ptpn11 gene, which regulates cell growth, differentiation and apoptosis via modulating various signaling pathways, such as the RAS/ERK signaling pathway, and participates in the PD-1/PD-L1 pathway governing immune surveillance. It has been recognized as a breakthrough antitumor therapeutic target. Besides, numerous studies have shown that SHP2 plays an important role in the regulation of inflammatory diseases. However, inhibitors targeting the active site of SHP2 lack drug-likeness due to their low selectivity and poor bioavailability, thus none has advanced to clinical development. Recently, allosteric inhibitors that stabilize the inactive conformation of SHP2 have achieved breakthrough progress, providing the clinical proof for the druggability of SHP2 as an antitumor drug target. This paper reviews the recently reported design and discovery of SHP2 small molecule inhibitors, focused on the structure-activity relationship (SAR) analysis of several representative SHP2 inhibitors, outlining the evolution and therapeutic potential of the small molecule inhibitors targeting SHP2.
Collapse
Affiliation(s)
- Jiao Kong
- Laboratory of Medicinal Chemical Biology, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College Suzhou 215123 China
| | - Ya-Qiu Long
- Laboratory of Medicinal Chemical Biology, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College Suzhou 215123 China
| |
Collapse
|
6
|
Zhao Z, Bai Y, Tian H, Shi B, Li X, Luo Y, Wang J, Hu J, Abbas Raza SH. Interference with ACSL1 gene in bovine adipocytes: Transcriptome profiling of circRNA related to unsaturated fatty acid production. Genomics 2021; 113:3967-3977. [PMID: 34601049 DOI: 10.1016/j.ygeno.2021.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 01/12/2023]
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) is a member of the acyl-CoA synthetase family that plays a vital role in lipid metabolism. We have previously shown that the ACSL1 gene regulates the composition of unsaturated fatty acids (UFAs) in bovine skeletal muscle, which in turn regulates the fatty acid synthesis and the generation of lipid droplets. Here, we used RNA-Seq to screen circRNAs that regulated the expression of ACSL1 gene and other UFA synthesis-related genes by RNA interference and noninterference in bovine adipocytes. The results of KEGG pathway analysis showed that the parental genes of differentially expressed (DE)-circRNAs were primarily enriched in the adipocytokine signaling pathway. The prediction results showed that novel_circ_0004855, novel_circ_0001507, novel_circ_0001731, novel_circ_0005276, novel_circ_0002060, novel_circ_0005405 and novel_circ_0004254 regulated UFA synthesis-related genes by interacting with the related miRNAs. These results could help expand our knowledge of the molecular mechanisms of circRNAs in the regulation of UFA synthesis in bovine adipocytes.
Collapse
Affiliation(s)
- Zhidong Zhao
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yanbin Bai
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Hongshan Tian
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Bingang Shi
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xupeng Li
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuzhu Luo
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiqing Wang
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiang Hu
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
7
|
Monda E, Rubino M, Lioncino M, Di Fraia F, Pacileo R, Verrillo F, Cirillo A, Caiazza M, Fusco A, Esposito A, Fimiani F, Palmiero G, Pacileo G, Calabrò P, Russo MG, Limongelli G. Hypertrophic Cardiomyopathy in Children: Pathophysiology, Diagnosis, and Treatment of Non-sarcomeric Causes. Front Pediatr 2021; 9:632293. [PMID: 33718303 PMCID: PMC7947260 DOI: 10.3389/fped.2021.632293] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a myocardial disease characterized by left ventricular hypertrophy not solely explained by abnormal loading conditions. Despite its rare prevalence in pediatric age, HCM carries a relevant risk of mortality and morbidity in both infants and children. Pediatric HCM is a large heterogeneous group of disorders. Other than mutations in sarcomeric genes, which represent the most important cause of HCM in adults, childhood HCM includes a high prevalence of non-sarcomeric causes, including inherited errors of metabolism (i.e., glycogen storage diseases, lysosomal storage diseases, and fatty acid oxidation disorders), malformation syndromes, neuromuscular diseases, and mitochondrial disease, which globally represent up to 35% of children with HCM. The age of presentation and the underlying etiology significantly impact the prognosis of children with HCM. Moreover, in recent years, different targeted approaches for non-sarcomeric etiologies of HCM have emerged. Therefore, the etiological diagnosis is a fundamental step in designing specific management and therapy in these subjects. The present review aims to provide an overview of the non-sarcomeric causes of HCM in children, focusing on the pathophysiology, clinical features, diagnosis, and treatment of these rare disorders.
Collapse
Affiliation(s)
- Emanuele Monda
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marta Rubino
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Lioncino
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Di Fraia
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Roberta Pacileo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Federica Verrillo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annapaola Cirillo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Martina Caiazza
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Adelaide Fusco
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Augusto Esposito
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabio Fimiani
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Palmiero
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Pacileo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Giovanna Russo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Institute of Cardiovascular Sciences, University College of London and St. Bartholomew's Hospital, London, United Kingdom
| |
Collapse
|
8
|
García-Gil M, Álvarez-Salafranca M, Valero-Torres A, Ara-Martín M. Melanoma in Noonan Syndrome With Multiple Lentigines (LEOPARD syndrome): A new case. ACTAS DERMO-SIFILIOGRAFICAS 2020. [DOI: 10.1016/j.adengl.2020.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
9
|
Alter S, Zimmer AD, Park M, Gong J, Caliebe A, Fölster-Holst R, Torrelo A, Colmenero I, Steinberg SF, Fischer J. Telangiectasia-ectodermal dysplasia-brachydactyly-cardiac anomaly syndrome is caused by de novo mutations in protein kinase D1. J Med Genet 2020; 58:415-421. [PMID: 32817298 DOI: 10.1136/jmedgenet-2019-106564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/18/2020] [Accepted: 05/30/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND We describe two unrelated patients who display similar clinical features including telangiectasia, ectodermal dysplasia, brachydactyly and congenital heart disease. METHODS We performed trio whole exome sequencing and functional analysis using in vitro kinase assays with recombinant proteins. RESULTS We identified two different de novo mutations in protein kinase D1 (PRKD1, NM_002742.2): c.1774G>C, p.(Gly592Arg) and c.1808G>A, p.(Arg603His), one in each patient. PRKD1 (PKD1, HGNC:9407) encodes a kinase that is a member of the protein kinase D (PKD) family of serine/threonine protein kinases involved in diverse cellular processes such as cell differentiation and proliferation and cell migration as well as vesicle transport and angiogenesis. Functional analysis using in vitro kinase assays with recombinant proteins showed that the mutation c.1808G>A, p.(Arg603His) represents a gain-of-function mutation encoding an enzyme with a constitutive, lipid-independent catalytic activity. The mutation c.1774G>C, p.(Gly592Arg) in contrast shows a defect in substrate phosphorylation representing a loss-of-function mutation. CONCLUSION The present cases represent a syndrome, which associates symptoms from several different organ systems: skin, teeth, bones and heart, caused by heterozygous de novo mutations in PRKD1 and expands the clinical spectrum of PRKD1 mutations, which have hitherto been linked to syndromic congenital heart disease and limb abnormalities.
Collapse
Affiliation(s)
- Svenja Alter
- Institute of Human Genetics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas David Zimmer
- Institute of Human Genetics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Misun Park
- Department of Pharmacology, Columbia University, New York, New York, USA
| | - Jianli Gong
- Department of Pharmacology, Columbia University, New York, New York, USA
| | - Almuth Caliebe
- Institute of Human Genetics, Christian-Albrechts University Kiel & University Hospital Schleswig-Holstein, Kiel, Germany
| | - Regina Fölster-Holst
- Department of Dermatology, Christian-Albrechts University Kiel & University Hospital Schleswig-Holstein, Kiel, Germany
| | - Antonio Torrelo
- Department of Dermatology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Isabel Colmenero
- Department of Pathology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York, New York, USA
| | - Judith Fischer
- Institute of Human Genetics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
García-Gil MF, Álvarez-Salafranca M, Valero-Torres A, Ara-Martín M. Melanoma in Noonan Syndrome With Multiple Lentigines (Leopard Syndrome): A New Case. ACTAS DERMO-SIFILIOGRAFICAS 2020; 111:619-621. [PMID: 32531240 DOI: 10.1016/j.ad.2019.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 01/01/2019] [Indexed: 11/28/2022] Open
Affiliation(s)
- M F García-Gil
- Servicio de Dermatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España.
| | - M Álvarez-Salafranca
- Servicio de Dermatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| | - A Valero-Torres
- Servicio de Anatomía Patológica, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| | - M Ara-Martín
- Servicio de Dermatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| |
Collapse
|
11
|
Ranza E, Guimier A, Verloes A, Capri Y, Marques C, Auclair M, Mathieu-Dramard M, Morin G, Thevenon J, Faivre L, Thauvin-Robinet C, Innes AM, Dyment DA, Vigouroux C, Amiel J. Overlapping phenotypes between SHORT and Noonan syndromes in patients with PTPN11 pathogenic variants. Clin Genet 2020; 98:10-18. [PMID: 32233106 DOI: 10.1111/cge.13746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
Overlapping syndromes such as Noonan, Cardio-Facio-Cutaneous, Noonan syndrome (NS) with multiple lentigines and Costello syndromes are genetically heterogeneous conditions sharing a dysregulation of the RAS/mitogen-activated protein kinase (MAPK) pathway and are known collectively as the RASopathies. PTPN11 was the first disease-causing gene identified in NS and remains the more prevalent. We report seven patients from three families presenting heterozygous missense variants in PTPN11 probably responsible for a disease phenotype distinct from the classical Noonan syndrome. The clinical presentation and common features of these seven cases overlap with the SHORT syndrome. The latter is the consequence of PI3K/AKT signaling deregulation with the predominant disease-causing gene being PIK3R1. Our data suggest that the phenotypic spectrum associated with pathogenic variants of PTPN11 could be wider than previously described, and this could be due to the dual activity of SHP2 (ie, PTPN11 gene product) on the RAS/MAPK and PI3K/AKT signaling.
Collapse
Affiliation(s)
- Emmanuelle Ranza
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Anne Guimier
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Alain Verloes
- Department of Genetics, APHP- Robert Debré University Hospital & INSERM UMR1141, Paris, France
| | - Yline Capri
- Department of Genetics, APHP- Robert Debré University Hospital & INSERM UMR1141, Paris, France
| | - Charles Marques
- Faculdade de Medicina, Centro Universitario Estacio, Ribeirao Preto, São Paulo, Brazil
| | - Martine Auclair
- Centre de Recherche Saint-Antoine, et Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM UMR_S 938, Paris, France
| | - Michèle Mathieu-Dramard
- Service de Génétique Clinique, Centre de référence maladies rares, CHU d'Amiens-site Sud, Amiens, France
| | - Gilles Morin
- Service de Génétique Clinique, Centre de référence maladies rares, CHU d'Amiens-site Sud, Amiens, France
| | - Julien Thevenon
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - Laurence Faivre
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David A Dyment
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Corinne Vigouroux
- Centre de Recherche Saint-Antoine, et Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM UMR_S 938, Paris, France.,AP-HP, Hôpital Saint-Antoine, Centre de Référence des Pathologies Rares de l'Insulino-Sécrétion et de l'Insulino-Sensibilité (PRISIS), Service d'Endocrinologie, Diabétologie et Endocrinologie de la Reproduction, and Laboratoire Commun de Biologie et Génétique Moléculaires, Paris, France
| | - Jeanne Amiel
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
12
|
Liu M, Jin HS, Park S. Protein and fat intake interacts with the haplotype of PTPN11_rs11066325, RPH3A_rs886477, and OAS3_rs2072134 to modulate serum HDL concentrations in middle-aged people. Clin Nutr 2020; 39:942-949. [PMID: 31006500 DOI: 10.1016/j.clnu.2019.03.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Low serum HDL cholesterol (HDL-C) concentration is a risk factor for cardiovascular diseases and it is influenced by genetic and environmental factors. We hypothesized that genetic variants that decrease serum HDL-C concentrations may interact with nutrient intakes in ways that increase or decrease the risk of cardiovascular disease. METHODS Candidate genetic variants that can lower serum HDL-C concentrations were explored by genome-wide association studies (GWAS), after adjusting for covariates, in the Ansan/Ansung cohort (n = 8842) from KoGES. The best genetic variants were selected and used to form a haplotype. According to the haplotype frequencies of SNPs, they were divided into major allele, heterozygote allele, and minor allele. The association of haplotype with serum HDL-C levels was determined using logistic regression after adjusting for confounding factors. Interaction of the haplotype with nutrient intake was also determined. RESULTS PTPN11_rs11066325, RPH3A_rs886477 and OAS3_rs2072134 were selected to modulate serum HDL-C levels from GWAS(P = 1.09E-09, 7.04E-10, and 1.27E-09, respectively). The adjusted odds ratios (ORs) for a decrease in serum HDL-C concentration in the minor-allele group of the haplotype were elevated by 1.534 fold, compared to the major-allele group of the haplotype. Furthermore, the adjusted ORs for serum LDL cholesterol and levels increased by 1.645 in the minor-alleles compared to the major-alleles of the haplotype without a significant change of serum cholesterol levels. Interestingly, the adjusted ORs for serum triglyceride were lower in the minor-alleles than in the major-alleles. The haplotype had a significant interaction with the intake of protein, fat, saturated fatty acids (SAF) and polyunsaturated fatty acids (PUFA; P < 0.05). In particular, the minor alleles of the haplotype decreased serum HDL-C levels compared to the major-alleles in the high intake of protein, fat, SFA, and PUFA, not in the low intake. CONCLUSIONS People carrying the minor-allele of haplotypes should avoid diets that are high in protein and fat, especially rich in SFA and PUFA.
Collapse
Affiliation(s)
- Meiling Liu
- Dept. of Food and Nutrition, Institue of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan, Chungnam, 31499, South Korea
| | - Hyun Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Chungnam, 31499, South Korea
| | - Sunmin Park
- Dept. of Food and Nutrition, Institue of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan, Chungnam, 31499, South Korea.
| |
Collapse
|
13
|
Lee CL, Tan LTHC, Lin HY, Hwu WL, Lee NC, Chien YH, Chuang CK, Wu MH, Wang JK, Chu SY, Lin JL, Lo FS, Su PH, Hsu CC, Ko YY, Chen MR, Chiu HC, Lin SP. Cardiac manifestations and gene mutations of patients with RASopathies in Taiwan. Am J Med Genet A 2019; 182:357-364. [PMID: 31837205 DOI: 10.1002/ajmg.a.61429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 11/07/2022]
Abstract
RASopathies are developmental diseases caused by mutations in rat sarcoma-mitogen-activated protein kinase pathway genes. These disorders, such as Noonan syndrome (NS) and NS-related disorders (NSRD), including cardio-facio-cutaneous (CFC) syndrome, Costello syndrome (CS), and NS with multiple lentigines (NSML; also known as LEOPARD syndrome), have a similar systemic phenotype. A wide spectrum of congenital heart disease and hypertrophic cardiomyopathy (HCMP) can exhibit major associated characteristics. A retrospective study was conducted at the Mackay Memorial Hospital, National Taiwan University Hospital, Buddhist Tzu-Chi General Hospital, Chang-Gung Memorial Hospital, Taichung Veterans General Hospital, and Chung Shan Medical University Hospital from January 2007 to December 2018. We reviewed the clinical records of 76 patients with a confirmed molecular diagnosis of RASopathies, including NS, CS, CFC syndrome, and NSML. We evaluated the demographic data and medical records with clinical phenotypes of cardiac structural anomalies using cross-sectional and color Doppler echocardiography, electrocardiographic findings, and follow-up data. A total of 47 (61.8%) patients had cardiac abnormalities. The prevalence of cardiac lesions according to each syndrome was 62.7, 50.0, 60.0, and 66.7% in patients with NS, CFC syndrome, CS, and NSML, respectively. An atrial septal defect was usually combined with other cardiac abnormalities, such as pulmonary stenosis (PS), HCMP, ventricular septal defect, or patent ductus arteriosus. Patients with NS most commonly showed PS. In patients with NSRD and cardiac abnormalities, HCMP (29.4%) was the most commonly observed cardiac lesion. PTPN11 was also the most frequently detected mutation in patients with NS and NSRD. Cardiac abnormalities were the most common symptoms observed in patients with RASopathies at the time of their first hospital visit. Performing precise analyses of genotype-cardiac phenotype correlations in a larger cohort will help us accurately diagnose RASopathy as soon as possible.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, Mackay Memorial Hospital, Hsinchu, Taiwan
| | | | - Hsiang-Yu Lin
- Department of Pediatrics and Rare Disease Center, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Nursing and Management, Mackay Junior College of Medicine, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Mei-Hwan Wu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jou-Kou Wang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shao-Yin Chu
- Department of Pediatrics, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan
| | - Ju-Li Lin
- Department of Pediatrics, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fu-Sung Lo
- Department of Pediatrics, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pen-Hua Su
- Department of Pediatrics, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Chi Hsu
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Yuan Ko
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Ren Chen
- Pediatric Cardiology Department, Mackay Children's Hospital, Taipei, Taiwan
| | - Hui-Ching Chiu
- Department of Pediatrics and Rare Disease Center, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics and Rare Disease Center, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
14
|
Cui H, Song L, Zhu C, Zhang C, Tang B, Wang S, Wu G, Zou Y, Huang X, Hui R, Wang S, Wang J. mTOR pathway in human cardiac hypertrophy caused by LEOPARD syndrome: a different role compared with animal models? Orphanet J Rare Dis 2019; 14:252. [PMID: 31722741 PMCID: PMC6854668 DOI: 10.1186/s13023-019-1204-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/13/2019] [Indexed: 11/10/2022] Open
Abstract
Background Animal studies suggested that blocking the activation of the mammalian target of rapamycin (mTOR) pathway might be effective to treat cardiac hypertrophy in LEOPARD syndrome (LS) caused by PTPN11 mutations. Results In the present study, mTOR pathway activity was examined in human myocardial samples from two patients with LS, four patients with hypertrophic cardiomyopathy (HCM), and four normal controls. The two patients with LS had p.Y279C and p.T468 M mutations of the PTPN11 gene, respectively. Although PTPN11 mutation showed initially positive regulation on phosphoinositide 3-kinase, overall the mTOR complex 1 pathway showed widely attenuated activity in LS. This included mildly hypophosphorylated mTOR and ribosomal protein S6 kinase and significantly hypophosphorylated Akt308 and ribosomal protein S6, which is similar to HCM. Akt473 is a basal molecule of the mTOR complex 2 pathway. Akt473 was less affected and showed hyperactivity in LS compared with HCM and normal controls. Additionally, MAPK/ERK kinase and ERK1/2 were significantly more phosphorylated in both HCM and LS than normal controls. Conclusions In LS, the mTOR signaling pathway shows similar activity to HCM and is attenuated compared with normal controls. Thus, caution should be applied when using rapamycin to treat heart hypertrophy in LS.
Collapse
Affiliation(s)
- Hao Cui
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Lei Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changsheng Zhu
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Ce Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Tang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Shengwei Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Guixin Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yubao Zou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Huang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
15
|
Eichhorn C, Voges I, Daubeney PEF. Out-of-hospital cardiac arrest and survival in a patient with Noonan syndrome and multiple lentigines: a case report. J Med Case Rep 2019; 13:194. [PMID: 31208451 PMCID: PMC6572739 DOI: 10.1186/s13256-019-2096-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/23/2019] [Indexed: 11/10/2022] Open
Abstract
Background A 9-year-old Arabic boy attending middle school presented with an out-of-hospital cardiac arrest due to ventricular fibrillation recorded by Holter electrocardiographic monitoring. He had a background history of Noonan syndrome with multiple lentigines (also known as LEOPARD syndrome), a rare condition of autosomal dominant inheritance with approximately 200 cases reported worldwide. Case presentation Apart from characteristic features, the boy was known to have asymmetric septal hypertrophy with a maximum wall thickness of 24 mm measured by cardiovascular magnetic resonance imaging. A day prior to the event, he attended cardiology follow-up at our institution, and Holter monitoring was commenced. Following cardiopulmonary resuscitation by bystanders and paramedics, he reverted back into sinus rhythm after a total downtime of 24 min. He was initially treated in the intensive care unit and underwent implantable cardioverter defibrillator implantation. He has made a full recovery and remains at the top of his class. Conclusion This case demonstrates that sudden cardiac arrest in patients with secondary forms of hypertrophic cardiomyopathy is not necessarily protected by apparently favorable phenotypes and that events may be preceded by non-sustained ventricular tachycardia observed by Holter monitoring. Implantable cardioverter defibrillator implantation plays a critical role in both primary and secondary prevention in patients at high risk of out-of-hospital cardiac arrest.
Collapse
Affiliation(s)
- Christian Eichhorn
- Department of Paediatric Cardiology, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Inga Voges
- Department of Paediatric Cardiology, Royal Brompton and Harefield NHS Foundation Trust, London, UK. .,Department of Congenital Heart Disease and Paediatric Cardiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| | - Piers E F Daubeney
- Department of Paediatric Cardiology, Royal Brompton and Harefield NHS Foundation Trust, London, UK.,National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
16
|
Abstract
More than any other organ, the heart is particularly sensitive to gene expression deregulation, often leading in the long run to impaired contractile performances and excessive fibrosis deposition progressing to heart failure. Recent investigations provide evidences that the protein phosphatases (PPs), as their counterpart protein kinases, are important regulators of cardiac physiology and development. Two main groups, the protein serine/threonine phosphatases and the protein tyrosine phosphatases (PTPs), constitute the PPs family. Here, we provide an overview of the role of PTP subfamily in the development of the heart and in cardiac pathophysiology. Based on recent in silico studies, we highlight the importance of PTPs as therapeutic targets for the development of new drugs to restore PTPs signaling in the early and late events of heart failure.
Collapse
Affiliation(s)
- Fallou Wade
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Centre, PO Box 3354, Riyadh, 11211, Saudi Arabia
| | - Karim Belhaj
- College of Medicine and Health Sciences, Al-Faisal University, Riyadh, 11211, Saudi Arabia
| | - Coralie Poizat
- Cardiovascular Research Program, King Faisal Specialist Hospital and Research Centre, PO Box 3354, Riyadh, 11211, Saudi Arabia. .,Biology Department, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
17
|
Abstract
RASopathies are a heterogeneous group of genetic syndromes characterized by mutations in genes that regulate cellular processes, including proliferation, differentiation, survival, migration, and metabolism. Excluding congenital heart defects, hypertrophic cardiomyopathy is the most frequent cardiovascular defect in patients affected by RASopathies. A worse outcome (in terms of surgical risk and/or mortality) has been described in a specific subset of Rasopathy patients with early onset, severe hypertrophic cardiomyopathy presenting with heart failure. New short-term therapy with a mammalian target of rapamycin inhibitor has recently been used to prevent heart failure in these patients with a severe form of hypertrophic cardiomyopathy.
Collapse
|
18
|
Zheng H, Yu WM, Waclaw RR, Kontaridis MI, Neel BG, Qu CK. Gain-of-function mutations in the gene encoding the tyrosine phosphatase SHP2 induce hydrocephalus in a catalytically dependent manner. Sci Signal 2018; 11:11/522/eaao1591. [PMID: 29559584 DOI: 10.1126/scisignal.aao1591] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Catalytically activating mutations in Ptpn11, which encodes the protein tyrosine phosphatase SHP2, cause 50% of Noonan syndrome (NS) cases, whereas inactivating mutations in Ptpn11 are responsible for nearly all cases of the similar, but distinct, developmental disorder Noonan syndrome with multiple lentigines (NSML; formerly called LEOPARD syndrome). However, both types of disease mutations are gain-of-function mutations because they cause SHP2 to constitutively adopt an open conformation. We found that the catalytic activity of SHP2 was required for the pathogenic effects of gain-of-function, disease-associated mutations on the development of hydrocephalus in the mouse. Targeted pan-neuronal knockin of a Ptpn11 allele encoding the active SHP2 E76K mutant resulted in hydrocephalus due to aberrant development of ependymal cells and their cilia. These pathogenic effects of the E76K mutation were suppressed by the additional mutation C459S, which abolished the catalytic activity of SHP2. Moreover, ependymal cells in NSML mice bearing the inactive SHP2 mutant Y279C were also unaffected. Mechanistically, the SHP2 E76K mutant induced developmental defects in ependymal cells by enhancing dephosphorylation and inhibition of the transcription activator STAT3. Whereas STAT3 activity was reduced in Ptpn11E76K/+ cells, the activities of the kinases ERK and AKT were enhanced, and neural cell-specific Stat3 knockout mice also manifested developmental defects in ependymal cells and cilia. These genetic and biochemical data demonstrate a catalytic-dependent role of SHP2 gain-of-function disease mutants in the pathogenesis of hydrocephalus.
Collapse
Affiliation(s)
- Hong Zheng
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wen-Mei Yu
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ronald R Waclaw
- Divisions of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maria I Kontaridis
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, New York University, New York, NY 10016, USA
| | - Cheng-Kui Qu
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
19
|
Dai Z, Whitt Z, Mighion LC, Pontoglio A, Bean LJ, Colombo R, Hegde M. Caution in interpretation of disease causality for heterozygous loss-of-function variants in the MYH8 gene associated with autosomal dominant disorder. Eur J Med Genet 2017; 60:312-316. [DOI: 10.1016/j.ejmg.2017.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 12/01/2022]
|
20
|
Cançado FHDSQ, da Silva LCP, Taitson PF, de Andrade ACDV, Pithon MM, Oliveira DD. Do you know this syndrome? Leopard syndrome. An Bras Dermatol 2017; 92:127-129. [PMID: 28225973 PMCID: PMC5312195 DOI: 10.1590/abd1806-4841.20174505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/20/2015] [Indexed: 11/22/2022] Open
Abstract
Hypertrophic cardiomyopathy is known as Leopard syndrome, which is a mnemonic rule for multiple lentigines (L), electrocardiographic conduction abnormalities (E), ocular hypertelorism (O), pulmonary stenosis (P), abnormalities of genitalia (A), retardation of growth (R), and deafness (D). We report the case of a 12-year-old patient with some of the abovementioned characteristics: hypertelorism, macroglossia, lentigines, hypospadias, cryptorchidism, subaortic stenosis, growth retardation, and hearing impairment. Due to this set of symptoms, we diagnosed Leopard syndrome.
Collapse
Affiliation(s)
| | - Luis Candido Pinto da Silva
- Odontology Department of the Pontifícia
Universidade Católica de Minas Gerais (PUC Minas) - Belo Horizonte (MG),
Brazil
| | - Paulo Franco Taitson
- Odontology Department of the Pontifícia
Universidade Católica de Minas Gerais (PUC Minas) - Belo Horizonte (MG),
Brazil
| | | | - Matheus Melo Pithon
- Health Department I of the Universidade Estadual do
Sudoeste da Bahia (UESB) - Itapetinga (BA), Brazil
| | - Dauro Douglas Oliveira
- Odontology Department of the Pontifícia
Universidade Católica de Minas Gerais (PUC Minas) - Belo Horizonte (MG),
Brazil
| |
Collapse
|
21
|
Jhang WK, Choi JH, Lee BH, Kim GH, Yoo HW. Cardiac Manifestations and Associations with Gene Mutations in Patients Diagnosed with RASopathies. Pediatr Cardiol 2016; 37:1539-1547. [PMID: 27554254 DOI: 10.1007/s00246-016-1468-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/16/2016] [Indexed: 01/09/2023]
Abstract
RASopathies are a group of syndromes caused by germline mutations of the RAS/MAPK pathway. They include Noonan syndrome, cardio-facio-cutaneous syndrome, Costello syndrome, and Noonan syndrome with multiple lentigines, which share many characteristic features including cardiac abnormalities. Here, we retrospectively reviewed the clinical manifestations and evaluated the genotype-phenotype associations with special focus on cardiac lesions of the patients with RASopathies. Cardiac symptoms were the most common initial presentation (27 %), except for admission to neonatal intensive care. Although there was a significant gap between the first visit to the hospital and the diagnosis of the genetic syndrome (19.9 ± 39.1 months), the age at the clinical diagnosis of the genetic syndrome was significantly lower in patients with CHD than in patients without CHD (47.26 ± 67.42 vs. 86.17 ± 85.66 months, p = 0.005). A wide spectrum of cardiac lesions was detected in 76.1 % (118/155) of included patients. The most common lesion was pulmonary stenosis, followed by atrial septal defect and hypertrophic cardiomyopathy (HCMP). About half of the pulmonary stenosis and HCMP patients progressed during the median follow-up period of 109.9 (range 9.7-315.4) months. Early rapid aggravation of cardiac lesions was linked to poor prognosis. MEK1, KRAS, and SOS1 mutations tend to be highly associated with pulmonary stenosis. Cardiologists may play important roles in early detection and diagnosis of RASopathies as well as associated CHDs. Due to the variety of clinical presentations and their progression of severity, proper management with regular long-term follow-up of these patients is essential.
Collapse
Affiliation(s)
- Won Kyoung Jhang
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jin-Ho Choi
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Medical Genetic Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Medical Genetic Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gu-Hwan Kim
- Medical Genetic Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Medical Genetic Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
22
|
|
23
|
Recent advances in RASopathies. J Hum Genet 2015; 61:33-9. [PMID: 26446362 DOI: 10.1038/jhg.2015.114] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/09/2015] [Accepted: 08/24/2015] [Indexed: 12/21/2022]
Abstract
RASopathies or RAS/mitogen-activated protein kinase (MAPK) syndromes are a group of phenotypically overlapping syndromes caused by germline mutations that encode components of the RAS/MAPK signaling pathway. These disorders include neurofibromatosis type I, Legius syndrome, Noonan syndrome, Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome), Costello syndrome, cardiofaciocutaneous (CFC) syndrome, Noonan-like syndrome, hereditary gingival fibromatosis and capillary malformation-arteriovenous malformation. Recently, novel gene variants, including RIT1, RRAS, RASA2, A2ML1, SOS2 and LZTR1, have been shown to be associated with RASopathies, further expanding the disease entity. Although further analysis will be needed, these findings will help to better elucidate an understanding of the pathogenesis of these disorders and will aid in the development of potential therapeutic approaches. In this review, we summarize the novel genes that have been reported to be associated with RASopathies and highlight the cardiovascular abnormalities that may arise in affected individuals.
Collapse
|
24
|
SHP2 sails from physiology to pathology. Eur J Med Genet 2015; 58:509-25. [PMID: 26341048 DOI: 10.1016/j.ejmg.2015.08.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/24/2015] [Accepted: 08/30/2015] [Indexed: 02/08/2023]
Abstract
Over the two past decades, mutations of the PTPN11 gene, encoding the ubiquitous protein tyrosine phosphatase SHP2 (SH2 domain-containing tyrosine phosphatase 2), have been identified as the causal factor of several developmental diseases (Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML), and metachondromatosis), and malignancies (juvenile myelomonocytic leukemia). SHP2 plays essential physiological functions in organism development and homeostasis maintenance by regulating fundamental intracellular signaling pathways in response to a wide range of growth factors and hormones, notably the pleiotropic Ras/Mitogen-Activated Protein Kinase (MAPK) and the Phosphoinositide-3 Kinase (PI3K)/AKT cascades. Analysis of the biochemical impacts of PTPN11 mutations first identified both loss-of-function and gain-of-function mutations, as well as more subtle defects, highlighting the major pathophysiological consequences of SHP2 dysregulation. Then, functional genetic studies provided insights into the molecular dysregulations that link SHP2 mutants to the development of specific traits of the diseases, paving the way for the design of specific therapies for affected patients. In this review, we first provide an overview of SHP2's structure and regulation, then describe its molecular roles, notably its functions in modulating the Ras/MAPK and PI3K/AKT signaling pathways, and its physiological roles in organism development and homeostasis. In the second part, we describe the different PTPN11 mutation-associated pathologies and their clinical manifestations, with particular focus on the biochemical and signaling outcomes of NS and NS-ML-associated mutations, and on the recent advances regarding the pathophysiology of these diseases.
Collapse
|
25
|
Hahn A, Lauriol J, Thul J, Behnke-Hall K, Logeswaran T, Schänzer A, Böğürcü N, Garvalov BK, Zenker M, Gelb BD, von Gerlach S, Kandolf R, Kontaridis MI, Schranz D. Rapidly progressive hypertrophic cardiomyopathy in an infant with Noonan syndrome with multiple lentigines: palliative treatment with a rapamycin analog. Am J Med Genet A 2015; 167A:744-51. [PMID: 25708222 DOI: 10.1002/ajmg.a.36982] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 01/02/2015] [Indexed: 12/16/2022]
Abstract
Noonan syndrome with multiple lentigines (NSML) frequently manifests with hypertrophic cardiomyopathy (HCM). Recently, it was demonstrated that mTOR inhibition reverses HCM in NSML mice. We report for the first time on the effects of treatment with a rapamycin analog in an infant with LS and malignant HCM. In the boy, progressive HCM was diagnosed during the first week of life and a diagnosis of NSML was established at age 20 weeks by showing a heterozygous Q510E mutation in PTPN11. Immunoblotting with antibodies against pERK, pAkt, and pS6RP in fibroblasts demonstrated enhanced Akt/mTOR pathway activity. Because of the patient's critical condition, everolimus therapy was started at age 24 weeks and continued until heart transplantation at age 36 weeks. Prior to surgery, heart failure improved from NYHA stage IV to II and brain natriuretic peptide values decreased from 9,600 to <1,000 pg/ml, but no reversal of cardiac hypertrophy was observed. Examination of the explanted heart revealed severe hypertrophy and myofiber disarray with extensive perivascular fibrosis. These findings provide evidence that Akt/mTOR activity is enhanced in NSML with HCM and suggest that rapamycin treatment could principally be feasible for infantile NSML. The preliminary experiences made in this single patient indicate that therapy should start early to prevent irreversible cardiac remodelling.
Collapse
Affiliation(s)
- Andreas Hahn
- Department of Child Neurology, University Hospital Giessen, Justus-Liebig University, Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mason SE, Lipshultz SE, Kaushal S, Fisher S. The implication of coronary artery malformations and congenital heart disease on cardiomyopathy. PROGRESS IN PEDIATRIC CARDIOLOGY 2014. [DOI: 10.1016/j.ppedcard.2014.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
27
|
|
28
|
Zhao S, Sedwick D, Wang Z. Genetic alterations of protein tyrosine phosphatases in human cancers. Oncogene 2014; 34:3885-94. [PMID: 25263441 PMCID: PMC4377308 DOI: 10.1038/onc.2014.326] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are enzymes that remove phosphate from tyrosine residues in proteins. Recent whole-exome sequencing of human cancer genomes reveals that many PTPs are frequently mutated in a variety of cancers. Among these mutated PTPs, protein tyrosine phosphatase T (PTPRT) appears to be the most frequently mutated PTP in human cancers. Beside PTPN11 which functions as an oncogene in leukemia, genetic and functional studies indicate that most of mutant PTPs are tumor suppressor genes. Identification of the substrates and corresponding kinases of the mutant PTPs may provide novel therapeutic targets for cancers harboring these mutant PTPs.
Collapse
Affiliation(s)
- S Zhao
- 1] Division of Gastroenterology and Hepatology and Shanghai Institution of Digestive Disease, Shanghai Jiao-Tong University School of Medicine Renji Hospital, Shanghai, China [2] Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA [3] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - D Sedwick
- 1] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA [2] Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Z Wang
- 1] Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA [2] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
29
|
The protein tyrosine phosphatase Shp2 is required for the generation of oligodendrocyte progenitor cells and myelination in the mouse telencephalon. J Neurosci 2014; 34:3767-78. [PMID: 24599474 DOI: 10.1523/jneurosci.3515-13.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The protein tyrosine phosphatase Shp2 (PTPN11) is crucial for normal brain development and has been implicated in dorsal telencephalic neuronal and astroglia cell fate decisions. However, its roles in the ventral telencephalon and during oligodendrogenesis in the telencephalon remain largely unknown. Shp2 gain-of-function (GOF) mutations are observed in Noonan syndrome, a type of RASopathy associated with multiple phenotypes, including cardiovascular, craniofacial, and neurocognitive abnormalities. To gain insight into requirements for Shp2 (LOF) and the impact of abnormal Shp2 GOF mutations, we used a Shp2 conditional mutant allele (LOF) and a cre inducible Shp2-Q79R GOF transgenic mouse in combination with Olig2(cre/+) mice to target embryonic ventral telencephalic progenitors and the oligodendrocyte lineage. In the absence of Shp2 (LOF), neuronal cell types originating from progenitors in the ventral telencephalon were generated, but oligodendrocyte progenitor cell (OPC) generation was severely impaired. Late embryonic and postnatal Shp2 cKOs showed defects in the generation of OPCs throughout the telencephalon and subsequent reductions in white matter myelination. Conversely, transgenic expression of the Shp2 GOF Noonan syndrome mutation resulted in elevated OPC numbers in the embryo and postnatal brain. Interestingly, expression of this mutation negatively influenced myelination as mice displayed abnormal myelination and fewer myelinated axons in the white matter despite elevated OPC numbers. Increased proliferating OPCs and elevated MAPK activity were also observed during oligodendrogenesis after expression of Shp2 GOF mutation. These results support the notion that appropriate Shp2 activity levels control the number as well as the differentiation of oligodendrocytes during development.
Collapse
|
30
|
Qiu W, Wang X, Romanov V, Hutchinson A, Lin A, Ruzanov M, Battaile KP, Pai EF, Neel BG, Chirgadze NY. Structural insights into Noonan/LEOPARD syndrome-related mutants of protein-tyrosine phosphatase SHP2 (PTPN11). BMC STRUCTURAL BIOLOGY 2014; 14:10. [PMID: 24628801 PMCID: PMC4007598 DOI: 10.1186/1472-6807-14-10] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 03/06/2014] [Indexed: 12/19/2022]
Abstract
Background The ubiquitous non-receptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) plays a key role in RAS/ERK signaling downstream of most, if not all growth factors, cytokines and integrins, although its major substrates remain controversial. Mutations in PTPN11 lead to several distinct human diseases. Germ-line PTPN11 mutations cause about 50% of Noonan Syndrome (NS), which is among the most common autosomal dominant disorders. LEOPARD Syndrome (LS) is an acronym for its major syndromic manifestations: multiple Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary stenosis, Abnormalities of genitalia, Retardation of growth, and sensorineural Deafness. Frequently, LS patients have hypertrophic cardiomyopathy, and they might also have an increased risk of neuroblastoma (NS) and acute myeloid leukemia (AML). Consistent with the distinct pathogenesis of NS and LS, different types of PTPN11 mutations cause these disorders. Results Although multiple studies have reported the biochemical and biological consequences of NS- and LS-associated PTPN11 mutations, their structural consequences have not been analyzed fully. Here we report the crystal structures of WT SHP2 and five NS/LS-associated SHP2 mutants. These findings enable direct structural comparisons of the local conformational changes caused by each mutation. Conclusions Our structural analysis agrees with, and provides additional mechanistic insight into, the previously reported catalytic properties of these mutants. The results of our research provide new information regarding the structure-function relationship of this medically important target, and should serve as a solid foundation for structure-based drug discovery programs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Benjamin G Neel
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, M5G 2C4, Canada.
| | | |
Collapse
|
31
|
Praetorius C, Sturm RA, Steingrimsson E. Sun-induced freckling: ephelides and solar lentigines. Pigment Cell Melanoma Res 2014; 27:339-50. [DOI: 10.1111/pcmr.12232] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Christian Praetorius
- Department of Biochemistry and Molecular Biology; Biomedical Center; Faculty of Medicine; University of Iceland; Reykjavik Iceland
| | - Richard A. Sturm
- Melanogenix Group; Institute for Molecular Bioscience; The University of Queensland; Brisbane Qld Australia
- Dermatology Research Centre; School of Medicine; The University of Queensland; Princess Alexandra Hospital; Brisbane Qld Australia
| | - Eirikur Steingrimsson
- Department of Biochemistry and Molecular Biology; Biomedical Center; Faculty of Medicine; University of Iceland; Reykjavik Iceland
| |
Collapse
|
32
|
Zanella F, Lyon RC, Sheikh F. Modeling heart disease in a dish: from somatic cells to disease-relevant cardiomyocytes. Trends Cardiovasc Med 2013; 24:32-44. [PMID: 24054750 DOI: 10.1016/j.tcm.2013.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/07/2013] [Accepted: 06/10/2013] [Indexed: 01/02/2023]
Abstract
A scientific milestone that has tremendously impacted the cardiac research field has been the discovery and establishment of human-induced pluripotent stem cells (hiPSC). Key to this discovery has been uncovering a viable path in generating human patient and disease-specific cardiac cells to dynamically model and study human cardiac diseases in an in vitro setting. Recent studies have demonstrated that hiPSC-derived cardiomyocytes can be used to model and recapitulate various known disease features in hearts of patient donors harboring genetic-based cardiac diseases. Experimental drugs have also been tested in this setting and shown to alleviate disease phenotypes in hiPSC-derived cardiomyocytes, further paving the way for therapeutic interventions for cardiac disease. Here, we review state-of-the-art methods to generate high-quality hiPSC and differentiate them towards cardiomyocytes as well as the full range of genetic-based cardiac diseases, which have been modeled using hiPSC. We also provide future perspectives on exploiting the potential of hiPSC to compliment existing studies and gain new insights into the mechanisms underlying cardiac disease.
Collapse
Affiliation(s)
- Fabian Zanella
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Robert C Lyon
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Farah Sheikh
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Martínez-Quintana E, Rodríguez-González F. RASopatías: del síndrome de Noonan al síndrome LEOPARD. Rev Esp Cardiol 2013. [DOI: 10.1016/j.recesp.2013.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
34
|
Martínez-Quintana E, Rodríguez-González F. RASopathies: from Noonan to LEOPARD syndrome. ACTA ACUST UNITED AC 2013; 66:756-7. [PMID: 24773692 DOI: 10.1016/j.rec.2013.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/07/2013] [Indexed: 01/27/2023]
Affiliation(s)
- Efrén Martínez-Quintana
- Servicio de Cardiología, Complejo Hospitalario Universitario Insular-Materno Infantil, Las Palmas de Gran Canaria, Spain.
| | - Fayna Rodríguez-González
- Servicio de Oftalmología, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
35
|
Jia ZF, Cao XY, Cao DH, Kong F, Kharbuja P, Jiang J. Polymorphisms of PTPN11 gene could influence serum lipid levels in a sex-specific pattern. Lipids Health Dis 2013; 12:72. [PMID: 23672255 PMCID: PMC3685535 DOI: 10.1186/1476-511x-12-72] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/10/2013] [Indexed: 01/18/2023] Open
Abstract
Background Previous studies have reported that different genotypes of PTPN11 gene (protein tyrosine phosphatase, non-receptor 11) were associated with different levels of serum lipids. The aim of this study was to explore the relationship between single nucleotide polymorphisms (SNPs) of PTPN11 and serum lipids in Northeast Chinese. Methods A total of 1003 subjects, 584 males and 419 females, were included in the study and their serum lipids were determined. Five htSNPs (rs2301756, rs12423190, rs12229892, rs7958372 and rs4767860) of PTPN11 gene were genotyped using TaqMan assay method. Results All of the five SNPs were in Hardy-Weinberg equilibrium. The male subjects had higher triglyceride (TG), higher low-density lipoprotein cholesterol (LDL-C) and lower high-density lipoprotein cholesterol (HDL-C) level than females. In males, rs4767860 was found to be associated with serum TG and total cholesterol (TC) levels and rs12229892 was associated with TC level. However, these significant associations could not be observed in females. In females, rs2301756 was found to be associated with TG and rs7958372 was associated with LDL-C level. Haplotype analysis showed that the GCGTG haplotype was associated with slightly higher TG level and ATGCG with higher TC level. Conclusions SNPs of PTPN11 may play a role in serum lipids in a sex-specific pattern. However, more studies are needed to confirm the conclusion and explore the underlying mechanism.
Collapse
Affiliation(s)
- Zhi-Fang Jia
- Division of Clinical Epidemiology, First Hospital of Jilin University, Changchun 130021, China
| | | | | | | | | | | |
Collapse
|