Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Circulation ; 140(3): 207-224, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31163979

ABSTRACT

BACKGROUND: More than 90% of individuals with Noonan syndrome (NS) with mutations clustered in the CR2 domain of RAF1 present with severe and often lethal hypertrophic cardiomyopathy (HCM). The signaling pathways by which NS RAF1 mutations promote HCM remain elusive, and so far, there is no known treatment for NS-associated HCM. METHODS: We used patient-derived RAF1S257L/+ and CRISPR-Cas9-generated isogenic control inducible pluripotent stem cell (iPSC)-derived cardiomyocytes to model NS RAF1-associated HCM and to further delineate the molecular mechanisms underlying the disease. RESULTS: We show that mutant iPSC-derived cardiomyocytes phenocopy the pathology seen in hearts of patients with NS by exhibiting hypertrophy and structural defects. Through pharmacological and genetic targeting, we identify 2 perturbed concomitant pathways that, together, mediate HCM in RAF1 mutant iPSC-derived cardiomyocytes. Hyperactivation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), but not extracellular regulated kinase 1/2, causes myofibrillar disarray, whereas the enlarged cardiomyocyte phenotype is a direct consequence of increased extracellular regulated kinase 5 (ERK5) signaling, a pathway not previously known to be involved in NS. RNA-sequencing reveals genes with abnormal expression in RAF1 mutant iPSC-derived cardiomyocytes and identifies subsets of genes dysregulated by aberrant MEK1/2 or ERK5 pathways that could contribute to the NS-associated HCM. CONCLUSIONS: Taken together, the results of our study identify the molecular mechanisms by which NS RAF1 mutations cause HCM and reveal downstream effectors that could serve as therapeutic targets for treatment of NS and perhaps other, more common, congenital HCM disorders.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Induced Pluripotent Stem Cells/physiology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Mitogen-Activated Protein Kinase 7/genetics , Noonan Syndrome/genetics , Proto-Oncogene Proteins c-raf/genetics , Adolescent , CRISPR-Cas Systems/physiology , Cardiomyopathy, Hypertrophic/metabolism , Cells, Cultured , Child , Female , HEK293 Cells , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mitogen-Activated Protein Kinase 7/metabolism , Myocytes, Cardiac/physiology , Noonan Syndrome/metabolism , Proto-Oncogene Proteins c-raf/metabolism
4.
Semin Cell Dev Biol ; 37: 73-81, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25256404

ABSTRACT

Congenital heart disease is the most common human developmental disorder, affecting ∼1:100 newborns, and is the primary cause of birth-defect related deaths worldwide. As a major regulator of receptor tyrosine kinase (RTK), cytokine and G-protein coupled receptor signaling, the non-receptor protein tyrosine phosphatase SHP2 plays a critical role in normal cardiac development and function. Indeed, SHP2 participates in a wide variety of cellular functions, including proliferation, survival, differentiation, migration, and cell-cell communication. Moreover, human activating and inactivating mutations of SHP2 are responsible for two related developmental disorders called Noonan and LEOPARD Syndromes, respectively, which are both characterized, in part, by congenital heart defects. Structural, enzymologic, biochemical, and SHP2 mouse model studies have together greatly enriched our knowledge of SHP2 and, as such, have also uncovered the diverse roles for SHP2 in cardiac development, including its contribution to progenitor cell specification, cardiac morphogenesis, and maturation of cardiac valves and myocardial chambers. By delineating the precise mechanisms by which SHP2 is involved in regulating these processes, we can begin to better understand the pathogenesis of cardiac disease and find more strategic and effective therapies for treatment of patients with congenital heart disorders.


Subject(s)
Heart Defects, Congenital/genetics , Myocardium/cytology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Cell Survival , Heart/embryology , Heart Defects, Congenital/metabolism , Humans , Mutation , Myocardium/metabolism , Stem Cells/cytology , Stem Cells/metabolism
5.
Circulation ; 125(24): 2993-3003, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22610965

ABSTRACT

BACKGROUND: Chronic ß-adrenergic receptor (ß-AR) overstimulation, a hallmark of heart failure, is associated with increased cardiac expression of matrix metalloproteinases (MMPs). MMP-1 has been shown to cleave and activate the protease-activated receptor 1 (PAR1) in noncardiac cells. In the present study, we hypothesized that ß-AR stimulation would result in MMP-dependent PAR1 transactivation in cardiac cells. METHODS AND RESULTS: ß-AR stimulation of neonatal rat ventricular myocytes (NRVMs) or cardiac fibroblasts with isoproterenol transduced with an alkaline phosphatase-tagged PAR1 elicited a significant increase in alkaline phosphatase-PAR1 cleavage. This isoproterenol-dependent cleavage was significantly reduced by the broad-spectrum MMP inhibitor GM6001. Importantly, specific MMP-13 inhibitors also decreased alkaline phosphatase-PAR1 cleavage in isoproterenol-stimulated NRVMs, as well as in NRVMs stimulated with conditioned medium from isoproterenol-stimulated cardiac fibroblasts. Moreover, we found that recombinant MMP-13 stimulation cleaved alkaline phosphatase-PAR1 in NRVMs at DPRS(42)↓(43)FLLRN. This also led to the activation of the ERK1/2 pathway through Gαq in NRVMs and via the Gαq/ErbB receptor pathways in cardiac fibroblasts. MMP-13 elicited similar levels of ERK1/2 activation but lower levels of generation of inositol phosphates in comparison to thrombin. Finally, we demonstrated that either PAR1 genetic ablation or pharmacological inhibition of MMP-13 prevented isoproterenol-dependent cardiac dysfunction in mice. CONCLUSIONS: In this study, we demonstrate that ß-AR stimulation leads to MMP-13 transactivation of PAR1 in both cardiac fibroblasts and cardiomyocytes and that this likely contributes to pathological activation of Gαq and ErbB receptor-dependent pathways in the heart. We propose that this mechanism may underlie the development of ß-AR overstimulation-dependent cardiac dysfunction.


Subject(s)
Matrix Metalloproteinase 13/physiology , Myocytes, Cardiac/metabolism , Receptor, PAR-1/metabolism , Receptors, Adrenergic, beta/physiology , Transcriptional Activation , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Male , Mice , Middle Aged , Oncogene Proteins v-erbB/physiology , Signal Transduction
6.
Stem Cell Res Ther ; 14(1): 266, 2023 09 23.
Article in English | MEDLINE | ID: mdl-37740238

ABSTRACT

BACKGROUND: Polymorphic ventricular tachycardia (PMVT) is a rare genetic disease associated with structurally normal hearts which in 8% of cases can lead to sudden cardiac death, typically exercise-induced. We previously showed a link between the RyR2-H29D mutation and a clinical phenotype of short-coupled PMVT at rest using patient-specific hiPSC-derived cardiomyocytes (hiPSC-CMs). In the present study, we evaluated the effects of clinical and experimental anti-arrhythmic drugs on the intracellular Ca2+ handling, contractile and molecular properties in PMVT hiPSC-CMs in order to model a personalized medicine approach in vitro. METHODS: Previously, a blood sample from a patient carrying the RyR2-H29D mutation was collected and reprogrammed into several clones of RyR2-H29D hiPSCs, and in addition we generated an isogenic control by reverting the RyR2-H29D mutation using CRIPSR/Cas9 technology. Here, we tested 4 drugs with anti-arrhythmic properties: propranolol, verapamil, flecainide, and the Rycal S107. We performed fluorescence confocal microscopy, video-image-based analyses and biochemical analyses to investigate the impact of these drugs on the functional and molecular features of the PMVT RyR2-H29D hiPSC-CMs. RESULTS: The voltage-dependent Ca2+ channel inhibitor verapamil did not prevent the aberrant release of sarcoplasmic reticulum (SR) Ca2+ in the RyR2-H29D hiPSC-CMs, whereas it was prevented by S107, flecainide or propranolol. Cardiac tissue comprised of RyR2-H29D hiPSC-CMs exhibited aberrant contractile properties that were largely prevented by S107, flecainide and propranolol. These 3 drugs also recovered synchronous contraction in RyR2-H29D cardiac tissue, while verapamil did not. At the biochemical level, S107 was the only drug able to restore calstabin2 binding to RyR2 as observed in the isogenic control. CONCLUSIONS: By testing 4 drugs on patient-specific PMVT hiPSC-CMs, we concluded that S107 and flecainide are the most potent molecules in terms of preventing the abnormal SR Ca2+ release and contractile properties in RyR2-H29D hiPSC-CMs, whereas the effect of propranolol is partial, and verapamil appears ineffective. In contrast with the 3 other drugs, S107 was able to prevent a major post-translational modification of RyR2-H29D mutant channels, the loss of calstabin2 binding to RyR2. Using patient-specific hiPSC and CRISPR/Cas9 technologies, we showed that S107 is the most efficient in vitro candidate for treating the short-coupled PMVT at rest.


Subject(s)
Calcium , Tachycardia, Ventricular , Humans , Myocytes, Cardiac , Flecainide/pharmacology , Propranolol/pharmacology , Propranolol/therapeutic use , Anti-Arrhythmia Agents , Precision Medicine , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/genetics , Verapamil/pharmacology , Verapamil/therapeutic use
7.
Science ; 379(6632): eabo3627, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36538032

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a rare and severe condition that follows benign COVID-19. We report autosomal recessive deficiencies of OAS1, OAS2, or RNASEL in five unrelated children with MIS-C. The cytosolic double-stranded RNA (dsRNA)-sensing OAS1 and OAS2 generate 2'-5'-linked oligoadenylates (2-5A) that activate the single-stranded RNA-degrading ribonuclease L (RNase L). Monocytic cell lines and primary myeloid cells with OAS1, OAS2, or RNase L deficiencies produce excessive amounts of inflammatory cytokines upon dsRNA or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulation. Exogenous 2-5A suppresses cytokine production in OAS1-deficient but not RNase L-deficient cells. Cytokine production in RNase L-deficient cells is impaired by MDA5 or RIG-I deficiency and abolished by mitochondrial antiviral-signaling protein (MAVS) deficiency. Recessive OAS-RNase L deficiencies in these patients unleash the production of SARS-CoV-2-triggered, MAVS-mediated inflammatory cytokines by mononuclear phagocytes, thereby underlying MIS-C.


Subject(s)
COVID-19 , Cytokines , Endoribonucleases , SARS-CoV-2 , Systemic Inflammatory Response Syndrome , Child , Humans , COVID-19/immunology , Cytokines/genetics , Cytokines/immunology , Endoribonucleases/genetics , Endoribonucleases/metabolism , RNA, Double-Stranded , SARS-CoV-2/genetics , Systemic Inflammatory Response Syndrome/genetics
8.
Arterioscler Thromb Vasc Biol ; 31(5): 1116-23, 2011 May.
Article in English | MEDLINE | ID: mdl-21330604

ABSTRACT

OBJECTIVE: Cyclophilin A (CyPA, encoded by Ppia) is a proinflammatory protein secreted in response to oxidative stress in mice and humans. We recently demonstrated that CyPA increased angiotensin II (Ang II)-induced reactive oxygen species (ROS) production in the aortas of apolipoprotein E (Apoe)-/- mice. In this study, we sought to evaluate the role of CyPA in Ang II-induced cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was not significantly different between Ppia+/+ and Ppia-/- mice infused with Ang II (1000 ng/min per kg for 4 weeks). Therefore, we investigated the effect of CyPA under conditions of high ROS and inflammation using the Apoe-/- mice. In contrast to Apoe-/- mice, Apoe-/-Ppia-/- mice exhibited significantly less Ang II-induced cardiac hypertrophy. Bone marrow cell transplantation showed that CyPA in cells intrinsic to the heart plays an important role in the cardiac hypertrophic response. Ang II-induced ROS production, cardiac fibroblast proliferation, and cardiac fibroblast migration were markedly decreased in Apoe-/-Ppia-/- cardiac fibroblasts. Furthermore, CyPA directly induced the hypertrophy of cultured neonatal cardiac myocytes. CONCLUSIONS: CyPA is required for Ang II-mediated cardiac hypertrophy by directly potentiating ROS production, stimulating the proliferation and migration of cardiac fibroblasts, and promoting cardiac myocyte hypertrophy.


Subject(s)
Apolipoproteins E/deficiency , Cardiomegaly/enzymology , Cyclophilin A/metabolism , Myocardium/enzymology , Angiotensin II , Animals , Animals, Newborn , Apolipoproteins E/genetics , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/immunology , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cell Communication , Cell Movement , Cell Proliferation , Cells, Cultured , Cyclophilin A/deficiency , Cyclophilin A/genetics , Disease Models, Animal , Fibroblasts/metabolism , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Time Factors
9.
Circ Res ; 104(1): 113-23, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19023134

ABSTRACT

By mimicking sympathetic stimulation in vivo, we previously reported that mice globally lacking serotonin 5-HT(2B) receptors did not develop isoproterenol-induced left ventricular hypertrophy. However, the exact cardiac cell type(s) expressing 5-HT(2B) receptors (cardiomyocytes versus noncardiomyocytes) involved in pathological heart hypertrophy was never addressed in vivo. We report here that mice expressing the 5-HT(2B) receptor solely in cardiomyocytes, like global 5-HT(2B) receptor-null mice, are resistant to isoproterenol-induced cardiac hypertrophy and dysfunction, as well as to isoproterenol-induced increases in cytokine plasma-levels. These data reveal a key role of noncardiomyocytes in isoproterenol-induced hypertrophy in vivo. Interestingly, we show that primary cultures of angiotensinogen null adult cardiac fibroblasts are releasing cytokines on stimulation with either angiotensin II or serotonin, but not in response to isoproterenol stimulation, demonstrating a critical role of angiotensinogen in adrenergic-dependent cytokine production. We then show a functional interdependence between AT(1)Rs and 5-HT(2B) receptors in fibroblasts by revealing a transinhibition mechanism that may involve heterodimeric receptor complexes. Both serotonin- and angiotensin II-dependent cytokine production occur via a Src/heparin-binding epidermal growth factor-dependent transactivation of epidermal growth factor receptors in cardiac fibroblasts, supporting a common signaling pathway. Finally, we demonstrate that 5-HT(2B) receptors are overexpressed in hearts from patients with congestive heart failure, this overexpression being positively correlated with cytokine and norepinephrine plasma levels. Collectively, these results reveal for the first time that interactions between AT(1) and 5-HT(2B) receptors coexpressed by noncardiomyocytes are limiting key events in adrenergic agonist-induced, angiotensin-dependent cardiac hypertrophy. Accordingly, antagonists of 5-HT(2B) receptors might represent novel therapeutics for sympathetic overstimulation-dependent heart failure.


Subject(s)
Fibroblasts/physiology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Myocardium/pathology , Receptor, Angiotensin, Type 1/physiology , Receptor, Serotonin, 5-HT2B/physiology , Adult , Angiotensin II/deficiency , Angiotensin II/physiology , Angiotensin II/toxicity , Animals , Cells, Cultured/metabolism , Cytokines/blood , Cytokines/metabolism , ErbB Receptors/physiology , Female , Fibroblasts/drug effects , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/pathology , Heparin-binding EGF-like Growth Factor , Humans , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/prevention & control , Intercellular Signaling Peptides and Proteins/physiology , Isoproterenol/toxicity , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Norepinephrine/physiology , Protein Interaction Mapping , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/therapeutic use , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/physiology
11.
Stem Cell Reports ; 16(9): 2274-2288, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34403650

ABSTRACT

Heart injury has been reported in up to 20% of COVID-19 patients, yet the cause of myocardial histopathology remains unknown. Here, using an established in vivo hamster model, we demonstrate that SARS-CoV-2 can be detected in cardiomyocytes of infected animals. Furthermore, we found damaged cardiomyocytes in hamsters and COVID-19 autopsy samples. To explore the mechanism, we show that both human pluripotent stem cell-derived cardiomyocytes (hPSC-derived CMs) and adult cardiomyocytes (CMs) can be productively infected by SARS-CoV-2, leading to secretion of the monocyte chemoattractant cytokine CCL2 and subsequent monocyte recruitment. Increased CCL2 expression and monocyte infiltration was also observed in the hearts of infected hamsters. Although infected CMs suffer damage, we find that the presence of macrophages significantly reduces SARS-CoV-2-infected CMs. Overall, our study provides direct evidence that SARS-CoV-2 infects CMs in vivo and suggests a mechanism of immune cell infiltration and histopathology in heart tissues of COVID-19 patients.


Subject(s)
COVID-19/pathology , Chemokine CCL2/metabolism , Heart Injuries/virology , Monocytes/immunology , Myocytes, Cardiac/metabolism , Animals , Cell Communication/physiology , Cell Line , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Humans , Macrophages/immunology , Male , Myocytes, Cardiac/virology , Pluripotent Stem Cells/cytology , Vero Cells
12.
Res Sq ; 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33236003

ABSTRACT

Heart injury has been reported in up to 20% of COVID-19 patients, yet the cause of myocardial histopathology remains unknown. In order to study the cause of myocardial pathology in COVID-19 patients, we used a hamster model to determine whether following infection SARS-CoV-2, the causative agent of COVID-19, can be detected in heart tissues. Here, we clearly demonstrate that viral RNA and nucleocapsid protein is present in cardiomyocytes in the hearts of infected hamsters. Interestingly, functional cardiomyocyte associated gene expression was decreased in infected hamster hearts, corresponding to an increase in reactive oxygen species (ROS). This data using an animal model was further validated using autopsy heart samples of COVID-19 patients. Moreover, we show that both human pluripotent stem cell-derived cardiomyocytes (hPSC-derived CMs) and adult cardiomyocytes (CMs) can be infected by SARS-CoV-2 and that CCL2 is secreted upon SARS-CoV-2 infection, leading to monocyte recruitment. Increased CCL2 expression and macrophage infiltration was also observed in the hearts of infected hamsters. Using single cell RNA-seq, we also show that macrophages are able to decrease SARS-CoV-2 infection of CMs. Overall, our study provides direct evidence that SARS-CoV-2 infects CMs in vivo and proposes a mechanism of immune-cell infiltration and pathology in heart tissue of COVID-19 patients.

13.
EBioMedicine ; 60: 103024, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32980690

ABSTRACT

BACKGROUND: While mutations in the cardiac type 2 ryanodine receptor (RyR2) have been linked to exercise-induced or catecholaminergic polymorphic ventricular tachycardia (CPVT), its association with polymorphic ventricular tachycardia (PMVT) occurring at rest is unclear. We aimed at constructing a patient-specific human-induced pluripotent stem cell (hiPSC) model of PMVT occurring at rest linked to a single point mutation in RyR2. METHODS: Blood samples were obtained from a patient with PMVT at rest due to a heterozygous RyR2-H29D mutation. Patient-specific hiPSCs were generated from the blood samples, and the hiPSC-derived cardiomyocytes (CMs) were generated via directed differentiation. Using CRIPSR/Cas9 technology, isogenic controls were generated by correcting the RyR2-H29D mutation. Using patch-clamp, fluorescent confocal microscopy and video-image-based analysis, the molecular and functional properties of RyR2-H29D hiPSCCMs and control hiPSCCMs were compared. FINDINGS: RyR2-H29D hiPSCCMs exhibit intracellular sarcoplasmic reticulum (SR) Ca2+ leak through RyR2 under physiological pacing. RyR2-H29D enhances the contribution of inositol 1,4,5-trisphosphate receptors to excitation-contraction coupling (ECC) that exacerbates abnormal Ca2+ release in RyR2-H29D hiPSCCMs. RyR2-H29D hiPSCCMs exhibit shorter action potentials, delayed afterdepolarizations, arrhythmias and aberrant contractile properties compared to isogenic controls. The RyR2-H29D mutation causes post-translational remodeling that is fully reversed with isogenic controls. INTERPRETATION: To conclude, in a model based on a RyR2 point mutation that is associated with short-coupled PMVT at rest, RyR2-H29D hiPSCCMs exhibited aberrant intracellular Ca2+ homeostasis, shortened action potentials, arrhythmias and abnormal contractile properties. FUNDING: French Muscular Dystrophy Association (AFM; project 16,073, MNM2 2012 and 20,225), "Fondation de la Recherche Médicale" (FRM; SPF20130526710), "Institut National pour la Santé et la Recherche Médicale" (INSERM), National Institutes of Health (ARM; R01 HL145473) and New York State Department of Health (NYSTEM C029156).


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/therapy , Alleles , CRISPR-Cas Systems , Calcium/metabolism , Calcium Signaling , Genotype , Homeostasis , Humans , Immunohistochemistry , Mutation , Protein Processing, Post-Translational , Stem Cell Transplantation , Tachycardia, Ventricular/etiology
14.
Cell Stem Cell ; 27(1): 125-136.e7, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32579880

ABSTRACT

SARS-CoV-2 has caused the COVID-19 pandemic. There is an urgent need for physiological models to study SARS-CoV-2 infection using human disease-relevant cells. COVID-19 pathophysiology includes respiratory failure but involves other organ systems including gut, liver, heart, and pancreas. We present an experimental platform comprised of cell and organoid derivatives from human pluripotent stem cells (hPSCs). A Spike-enabled pseudo-entry virus infects pancreatic endocrine cells, liver organoids, cardiomyocytes, and dopaminergic neurons. Recent clinical studies show a strong association with COVID-19 and diabetes. We find that human pancreatic beta cells and liver organoids are highly permissive to SARS-CoV-2 infection, further validated using adult primary human islets and adult hepatocyte and cholangiocyte organoids. SARS-CoV-2 infection caused striking expression of chemokines, as also seen in primary human COVID-19 pulmonary autopsy samples. hPSC-derived cells/organoids provide valuable models for understanding the cellular responses of human tissues to SARS-CoV-2 infection and for disease modeling of COVID-19.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/virology , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Organoids/virology , Pneumonia, Viral/virology , Tropism , Angiotensin-Converting Enzyme 2 , Animals , Autopsy , COVID-19 , Cell Line , Coronavirus Infections/pathology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Induced Pluripotent Stem Cells/virology , Liver/pathology , Mice , Pancreas/pathology , Pancreas/virology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , SARS-CoV-2 , Virus Internalization
15.
Stem Cell Res ; 34: 101374, 2019 01.
Article in English | MEDLINE | ID: mdl-30640061

ABSTRACT

Noonan syndrome with multiple lentigines (NSML), formerly known as LEOPARD Syndrome, is a rare autosomal dominant disorder. Approximately 90% of NSML cases are caused by missense mutations in the PTPN11 gene which encodes the protein tyrosine phosphatase SHP2. A human induced pluripotent stem cell (iPSC) line was generated using peripheral blood mononuclear cells (PBMCs) from a patient with NSML that carries a gene mutation of p.Q510P on the PTPN11 gene using non-integrating Sendai virus technique. This iPSC line offers a useful resource to study the disease pathophysiology and a cell-based model for drug development to treat NSML.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/pathology , LEOPARD Syndrome/genetics , LEOPARD Syndrome/pathology , Mutation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Adolescent , Base Sequence , Cell Line , Female , Humans
17.
Circulation ; 107(25): 3223-9, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12810613

ABSTRACT

BACKGROUND: Identification of factors regulating myocardial structure and function is important to understand the pathogenesis of heart disease. We previously reported that 5-HT2B receptor ablation in mice leads to dilated cardiomyopathy. In this study, we investigated the pathological consequence of overexpressing 5-HT2B receptors in heart in vivo. METHODS AND RESULTS: We have generated transgenic mice overexpressing the Gq-coupled 5-HT2B receptor specifically in heart. We found that overexpression of 5-HT2B receptor in heart leads to ventricular hypertrophy as the result of increased cell number and size. Increased atrial natriuretic peptide and myosin heavy chain expression demonstrated activation of the molecular program for cardiac hypertrophy. Echocardiographic analysis indicated the presence of thickened ventricular free wall without alteration of the systolic function, showing that transgenic mice have compensated hypertrophy. Electron microscopic analysis revealed structural abnormalities including mitochondrial proliferation, as also manifested by histological staining. Transgenic mouse heart displayed a specific reduction in the expression levels of the adenine nucleotide translocator associated to increase in the succinate dehydrogenase and cytochrome C oxidase mitochondrial activities. CONCLUSIONS: Our results constitute the first genetic evidence that overexpression of the 5-HT2B receptor in the heart leads to compensated hypertrophic cardiomyopathy associated with proliferation of the mitochondria. This observation suggests a role for mitochondria in the hypertrophic signaling that is regulated by serotonin. These transgenic mice provide a new genetic model for hypertrophic heart disease.


Subject(s)
Cardiomegaly/physiopathology , Mitochondria/metabolism , Myocardium/metabolism , Receptors, Serotonin/biosynthesis , Animals , Atrial Natriuretic Factor/metabolism , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Cell Count , Cell Size , Disease Models, Animal , Echocardiography , Electron Transport Complex IV/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression , Heart Ventricles/pathology , Heterotrimeric GTP-Binding Proteins/metabolism , Male , Mice , Mice, Transgenic , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial ADP, ATP Translocases/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Myosin Heavy Chains/metabolism , Organ Specificity , Receptor, Serotonin, 5-HT2B , Receptors, Serotonin/genetics , Signal Transduction , Succinate Dehydrogenase/metabolism
18.
Circulation ; 110(8): 969-74, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15302781

ABSTRACT

BACKGROUND: The serotonergic 5-HT2B receptor regulates cardiomyocyte development and growth. A putative contribution of this receptor to fibroblast-dependent cardiac function has not been identified. METHODS AND RESULTS: By mimicking sympathetic stimulation with chronic isoproterenol perfusion in vivo, we found that mice developed a cardiac hypertrophy, which was prevented by exposure to the 5-HT2B receptor antagonists SB206553 or SB215505 or in 5-HT2B receptor-knockout mice. The isoproterenol-induced hypertrophy was associated with an increase in the plasma levels of interleukin-1beta and tumor necrosis factor-alpha but not interleukin-6. In contrast, the plasma isoproterenol-induced cytokine increase was not observed in either 5-HT2B receptor-mutant or wild-type mice perfused with isoproterenol+SB206553. We demonstrated that stimulation of wild-type cardiac fibroblasts by isoproterenol markedly increased the production of the interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha cytokines. Strikingly, we found that this isoproterenol-induced cytokine production was abolished by SB206553 or in 5-HT2B receptor-knockout fibroblasts. Serotonin also stimulated production of the 3 cytokines in wild-type fibroblasts, which was effectively reduced in 5-HT2B receptor-knockout fibroblasts. CONCLUSIONS: Our results demonstrate for the first time that 5-HT2B receptors are essential for isoproterenol-induced cardiac hypertrophy, which involves the regulation of interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha cytokine production by cardiac fibroblasts.


Subject(s)
Cardiomegaly/physiopathology , Fibroblasts/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Myocytes, Cardiac/cytology , Pyridines/therapeutic use , Quinolines/pharmacology , Receptor, Serotonin, 5-HT2B/physiology , Serotonin Antagonists/therapeutic use , Sympathetic Nervous System/physiopathology , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Antagonists , Adrenergic beta-Agonists/toxicity , Animals , Cardiomegaly/chemically induced , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heart Ventricles/cytology , Imidazoles/pharmacology , Interleukin-1/biosynthesis , Interleukin-1/blood , Interleukin-1/genetics , Interleukin-6/biosynthesis , Interleukin-6/blood , Interleukin-6/genetics , Isoproterenol/toxicity , Mice , Mice, Knockout , Propanolamines/pharmacology , Pyridines/pharmacology , Quinolines/therapeutic use , Receptor, Serotonin, 5-HT2B/deficiency , Receptor, Serotonin, 5-HT2B/genetics , Receptors, Adrenergic, beta-1/analysis , Receptors, Adrenergic, beta-2/analysis , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Sympathetic Nervous System/drug effects , Sympathomimetics/toxicity , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
19.
Sci Signal ; 7(348): ra100, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25336613

ABSTRACT

The Ras-related guanosine triphosphatase RhoA mediates pathological cardiac hypertrophy, but also promotes cell survival and is cardioprotective after ischemia/reperfusion injury. To understand how RhoA mediates these opposing roles in the myocardium, we generated mice with a cardiomyocyte-specific deletion of RhoA. Under normal conditions, the hearts from these mice showed functional, structural, and growth parameters similar to control mice. Additionally, the hearts of the cardiomyocyte-specific, RhoA-deficient mice subjected to transverse aortic constriction (TAC)-a procedure that induces pressure overload and, if prolonged, heart failure-exhibited a similar amount of hypertrophy as those of the wild-type mice subjected to TAC. Thus, neither normal cardiac homeostasis nor the initiation of compensatory hypertrophy required RhoA in cardiomyocytes. However, in response to chronic TAC, hearts from mice with cardiomyocyte-specific deletion of RhoA showed greater dilation, with thinner ventricular walls and larger chamber dimensions, and more impaired contractile function than those from control mice subjected to chronic TAC. These effects were associated with aberrant calcium signaling, as well as decreased activity of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and AKT. In addition, hearts from mice with cardiomyocyte-specific RhoA deficiency also showed less fibrosis in response to chronic TAC, with decreased transcriptional activation of genes involved in fibrosis, including myocardin response transcription factor (MRTF) and serum response factor (SRF), suggesting that the fibrotic response to stress in the heart depends on cardiomyocyte-specific RhoA signaling. Our data indicated that RhoA regulates multiple pathways in cardiomyocytes, mediating both cardioprotective (hypertrophy without dilation) and cardio-deleterious effects (fibrosis).


Subject(s)
Endomyocardial Fibrosis/enzymology , Heart Failure/enzymology , MAP Kinase Signaling System , Myocytes, Cardiac/enzymology , Stress, Physiological , rho GTP-Binding Proteins/metabolism , Animals , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Heart Failure/genetics , Heart Failure/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
20.
Nat Med ; 17(12): 1668-73, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22120177

ABSTRACT

Tissue homeostasis requires an effective, limited wound-healing response to injury. In chronic disease, failure to regenerate parenchymal tissue leads to the replacement of lost cellular mass with a fibrotic matrix. The mechanisms that dictate the balance of cell regeneration and fibrogenesis are not well understood. Here we report that fibrogenic hepatic stellate cells (HSCs) in the liver are negative regulators of hepatocyte regeneration. This negative regulatory function requires stimulation of the 5-hydroxytryptamine 2B receptor (5-HT(2B)) on HSCs by serotonin, which activates expression of transforming growth factor ß1 (TGF-ß1), a powerful suppressor of hepatocyte proliferation, through signaling by mitogen-activated protein kinase 1 (ERK) and the transcription factor JunD. Selective antagonism of 5-HT(2B) enhanced hepatocyte growth in models of acute and chronic liver injury. We also observed similar effects in mice lacking 5-HT(2B) or JunD or upon selective depletion of HSCs in wild-type mice. Antagonism of 5-HT(2B) attenuated fibrogenesis and improved liver function in disease models in which fibrosis was pre-established and progressive. Pharmacological targeting of 5-HT(2B) is clinically safe in humans and may be therapeutic in chronic liver disease.


Subject(s)
Liver Cirrhosis/therapy , Receptor, Serotonin, 5-HT2B/metabolism , Wound Healing , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chronic Disease , Electrophoresis, Polyacrylamide Gel , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Immunohistochemistry , Indoles/pharmacology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2B/drug effects , Serotonin/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Urea/analogs & derivatives , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL