Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3595, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678016

ABSTRACT

Plasticity among cell lineages is a fundamental, but poorly understood, property of regenerative tissues. In the gut tube, the small intestine absorbs nutrients, whereas the colon absorbs electrolytes. In a striking display of inherent plasticity, adult colonic mucosa lacking the chromatin factor SATB2 is converted to small intestine. Using proteomics and CRISPR-Cas9 screening, we identify MTA2 as a crucial component of the molecular machinery that, together with SATB2, restrains colonic plasticity. MTA2 loss in the adult mouse colon activated lipid absorptive genes and functional lipid uptake. Mechanistically, MTA2 co-occupies DNA with HNF4A, an activating pan-intestinal transcription factor (TF), on colonic chromatin. MTA2 loss leads to HNF4A release from colonic chromatin, and accumulation on small intestinal chromatin. SATB2 similarly restrains colonic plasticity through an HNF4A-dependent mechanism. Our study provides a generalizable model of lineage plasticity in which broadly-expressed TFs are retained on tissue-specific enhancers to maintain cell identity and prevent activation of alternative lineages, and their release unleashes plasticity.


Subject(s)
Chromatin , Colon , Hepatocyte Nuclear Factor 4 , Intestine, Small , Matrix Attachment Region Binding Proteins , Animals , Hepatocyte Nuclear Factor 4/metabolism , Hepatocyte Nuclear Factor 4/genetics , Intestine, Small/metabolism , Colon/metabolism , Mice , Chromatin/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Male , Cell Plasticity/genetics , Cell Lineage , Mice, Knockout
2.
bioRxiv ; 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37034724

ABSTRACT

Transition between activation and quiescence programs in hematopoietic stem and progenitor cells (HSC/HSPCs) is perceived to be governed intrinsically and by microenvironmental co-adaptation. However, HSC programs dictating both transition and adaptability, remain poorly defined. Single cell multiome analysis divulging differential transcriptional activity between distinct HSPC states, indicated for the exclusive absence of Fli-1 motif from quiescent HSCs. We reveal that Fli-1 activity is essential for HSCs during regenerative hematopoiesis. Fli-1 directs activation programs while manipulating cellular sensory and output machineries, enabling HSPCs co-adoptability with a stimulated vascular niche. During regenerative conditions, Fli-1 presets and enables propagation of niche-derived Notch1 signaling. Constitutively induced Notch1 signaling is sufficient to recuperate functional HSC impairments in the absence of Fli-1. Applying FLI-1 modified-mRNA transduction into lethargic adult human mobilized HSPCs, enables their vigorous niche-mediated expansion along with superior engraftment capacities. Thus, decryption of stem cell activation programs offers valuable insights for immune regenerative medicine.

3.
Int J Mol Sci ; 23(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35008993

ABSTRACT

Cell migration is a complex, tightly regulated multistep process in which cytoskeletal reorganization and focal adhesion redistribution play a central role. Core to both individual and collective migration is the persistent random walk, which is characterized by random force generation and resistance to directional change. We first discuss a model that describes the stochastic movement of ECs and characterizes EC persistence in wound healing. To that end, we pharmacologically disrupted cytoskeletal dynamics, cytochalasin D for actin and nocodazole for tubulin, to understand its contributions to cell morphology, stiffness, and motility. As such, the use of Atomic Force Microscopy (AFM) enabled us to probe the topography and stiffness of ECs, while time lapse microscopy provided observations in wound healing models. Our results suggest that actin and tubulin dynamics contribute to EC shape, compressive moduli, and directional organization in collective migration. Insights from the model and time lapse experiment suggest that EC speed and persistence are directionally organized in wound healing. Pharmacological disruptions suggest that actin and tubulin dynamics play a role in collective migration. Current insights from both the model and experiment represent an important step in understanding the biomechanics of EC migration as a therapeutic target.


Subject(s)
Cell Tracking , Cytoskeleton/metabolism , Endothelial Cells/physiology , Algorithms , Animals , Biomarkers , Cell Movement , Cell Tracking/methods , Fluorescent Antibody Technique , Humans , Microscopy, Atomic Force , Models, Biological , Molecular Imaging/methods , Wound Healing
4.
Nat Cardiovasc Res ; 1: 882-899, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36713285

ABSTRACT

Current dogma dictates that during adulthood, endothelial cells (ECs) are locked in an immutable stable homeostatic state. By contrast, herein we show that maintenance of EC fate and function are linked and active processes, which depend on the constitutive cooperativity of only two ETS-transcription factors (TFs) ERG and Fli1. While deletion of either Fli1 or ERG manifest subtle vascular dysfunction, their combined genetic deletion in adult EC results in acute vasculopathy and multiorgan failure, due to loss of EC fate and integrity, hyperinflammation, and spontaneous thrombosis, leading to death. ERG and Fli1 co-deficiency cause rapid transcriptional silencing of pan- and organotypic vascular core genes, with dysregulation of inflammation and coagulation pathways. Vascular hyperinflammation leads to impaired hematopoiesis with myeloid skewing. Accordingly, enforced ERG and FLI1 expression in adult human mesenchymal stromal cells activates vascular programs and functionality enabling engraftment of perfusable vascular network. GWAS-analysis identified vascular diseases are associated with FLI1/Erg mutations. Constitutive expression of ERG and Fli1 uphold EC fate, physiological function, and resilience in adult vasculature; while their functional loss can contribute to systemic human diseases.

5.
Dev Cell ; 56(22): 3042-3051, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34813766

ABSTRACT

Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.


Subject(s)
Endothelial Cells/cytology , Kidney/blood supply , Microvessels/cytology , Neovascularization, Physiologic/physiology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Humans , Kidney/cytology
6.
Nature ; 585(7825): 426-432, 2020 09.
Article in English | MEDLINE | ID: mdl-32908310

ABSTRACT

Endothelial cells adopt tissue-specific characteristics to instruct organ development and regeneration1,2. This adaptability is lost in cultured adult endothelial cells, which do not vascularize tissues in an organotypic manner. Here, we show that transient reactivation of the embryonic-restricted ETS variant transcription factor 2 (ETV2)3 in mature human endothelial cells cultured in a serum-free three-dimensional matrix composed of a mixture of laminin, entactin and type-IV collagen (LEC matrix) 'resets' these endothelial cells to adaptable, vasculogenic cells, which form perfusable and plastic vascular plexi. Through chromatin remodelling, ETV2 induces tubulogenic pathways, including the activation of RAP1, which promotes the formation of durable lumens4,5. In three-dimensional matrices-which do not have the constraints of bioprinted scaffolds-the 'reset' vascular endothelial cells (R-VECs) self-assemble into stable, multilayered and branching vascular networks within scalable microfluidic chambers, which are capable of transporting human blood. In vivo, R-VECs implanted subcutaneously in mice self-organize into durable pericyte-coated vessels that functionally anastomose to the host circulation and exhibit long-lasting patterning, with no evidence of malformations or angiomas. R-VECs directly interact with cells within three-dimensional co-cultured organoids, removing the need for the restrictive synthetic semipermeable membranes that are required for organ-on-chip systems, therefore providing a physiological platform for vascularization, which we call 'Organ-On-VascularNet'. R-VECs enable perfusion of glucose-responsive insulin-secreting human pancreatic islets, vascularize decellularized rat intestines and arborize healthy or cancerous human colon organoids. Using single-cell RNA sequencing and epigenetic profiling, we demonstrate that R-VECs establish an adaptive vascular niche that differentially adjusts and conforms to organoids and tumoroids in a tissue-specific manner. Our Organ-On-VascularNet model will permit metabolic, immunological and physiochemical studies and screens to decipher the crosstalk between organotypic endothelial cells and parenchymal cells for identification of determinants of endothelial cell heterogeneity, and could lead to advances in therapeutic organ repair and tumour targeting.


Subject(s)
Blood Vessels/cytology , Carcinogenesis , Endothelial Cells/cytology , Hemodynamics , Neoplasms/blood supply , Organogenesis , Organoids/blood supply , Blood Vessels/growth & development , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Chromatin/metabolism , Epigenesis, Genetic , Epigenomics , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Islets of Langerhans/blood supply , Models, Biological , Organ Specificity , RNA-Seq , Single-Cell Analysis , Transcription Factors , Transcriptome
7.
Nat Commun ; 10(1): 5705, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836710

ABSTRACT

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Subject(s)
Capillaries/growth & development , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Kidney Glomerulus/blood supply , Animals , Capillaries/cytology , Capillaries/metabolism , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Kidney Glomerulus/growth & development , Kidney Glomerulus/metabolism , Male , Mice , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
8.
Circulation ; 140(14): 1188-1204, 2019 10.
Article in English | MEDLINE | ID: mdl-31567019

ABSTRACT

BACKGROUND: Arrhythmogenic cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium, resulting in heart failure and sudden cardiac death. The most aggressive arrhythmogenic cardiomyopathy/ARVC subtype is ARVC type 5 (ARVC5), caused by a p.S358L mutation in TMEM43 (transmembrane protein 43). The function and localization of TMEM43 are unknown, as is the mechanism by which the p.S358L mutation causes the disease. Here, we report the characterization of the first transgenic mouse model of ARVC5. METHODS: We generated transgenic mice overexpressing TMEM43 in either its wild-type or p.S358L mutant (TMEM43-S358L) form in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. RESULTS: We found that mice expressing TMEM43-S358L recapitulate the human disease and die at a young age. Mutant TMEM43 causes cardiomyocyte death and severe fibrofatty replacement. We also demonstrate that TMEM43 localizes at the nuclear membrane and interacts with emerin and ß-actin. TMEM43-S358L shows partial delocalization to the cytoplasm, reduced interaction with emerin and ß-actin, and activation of glycogen synthase kinase-3ß (GSK3ß). Furthermore, we show that targeting cardiac fibrosis has no beneficial effect, whereas overexpression of the calcineurin splice variant calcineurin Aß1 results in GSK3ß inhibition and improved cardiac function and survival. Similarly, treatment of TMEM43 mutant mice with a GSK3ß inhibitor improves cardiac function. Finally, human induced pluripotent stem cells bearing the p.S358L mutation also showed contractile dysfunction that was partially restored after GSK3ß inhibition. CONCLUSIONS: Our data provide evidence that TMEM43-S358L leads to sustained cardiomyocyte death and fibrofatty replacement. Overexpression of calcineurin Aß1 in TMEM43 mutant mice or chemical GSK3ß inhibition improves cardiac function and increases mice life span. Our results pave the way toward new therapeutic approaches for ARVC5.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Ventricular Dysfunction/pathology , Animals , Calcineurin/genetics , Calcineurin/metabolism , Cell Differentiation , Cell Survival/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Heart Ventricles/physiopathology , Humans , Induced Pluripotent Stem Cells/cytology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Severity of Illness Index , Ventricular Dysfunction/mortality
9.
Circ Res ; 125(2): 170-183, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31145021

ABSTRACT

RATIONALE: RBPs (RNA binding proteins) play critical roles in the cell by regulating mRNA transport, splicing, editing, and stability. The RBP SRSF3 (serine/arginine-rich splicing factor 3) is essential for blastocyst formation and for proper liver development and function. However, its role in the heart has not been explored. OBJECTIVE: To investigate the role of SRSF3 in cardiac function. METHODS AND RESULTS: Cardiac SRSF3 expression was high at mid gestation and decreased during late embryonic development. Mice lacking SRSF3 in the embryonic heart showed impaired cardiomyocyte proliferation and died in utero. In the adult heart, SRSF3 expression was reduced after myocardial infarction, suggesting a possible role in cardiac homeostasis. To determine the role of this RBP in the adult heart, we used an inducible, cardiomyocyte-specific SRSF3 knockout mouse model. After SRSF3 depletion in cardiomyocytes, mice developed severe systolic dysfunction that resulted in death within 8 days. RNA-Seq analysis revealed downregulation of mRNAs encoding sarcomeric and calcium handling proteins. Cardiomyocyte-specific SRSF3 knockout mice also showed evidence of alternative splicing of mTOR (mammalian target of rapamycin) mRNA, generating a shorter protein isoform lacking catalytic activity. This was associated with decreased phosphorylation of 4E-BP1 (eIF4E-binding protein 1), a protein that binds to eIF4E (eukaryotic translation initiation factor 4E) and prevents mRNA decapping. Consequently, we found increased decapping of mRNAs encoding proteins involved in cardiac contraction. Decapping was partially reversed by mTOR activation. CONCLUSIONS: We show that cardiomyocyte-specific loss of SRSF3 expression results in decapping of critical mRNAs involved in cardiac contraction. The molecular mechanism underlying this effect likely involves the generation of a short mTOR isoform by alternative splicing, resulting in reduced 4E-BP1 phosphorylation. The identification of mRNA decapping as a mechanism of systolic heart failure may open the way to the development of urgently needed therapeutic tools.


Subject(s)
Myocytes, Cardiac/metabolism , Serine-Arginine Splicing Factors/genetics , Ventricular Dysfunction/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/physiology , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors/metabolism , Systole , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Ventricular Dysfunction/metabolism
12.
J Am Coll Cardiol ; 71(6): 654-667, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29420962

ABSTRACT

BACKGROUND: In response to pressure overload, the heart develops ventricular hypertrophy that progressively decompensates and leads to heart failure. This pathological hypertrophy is mediated, among others, by the phosphatase calcineurin and is characterized by metabolic changes that impair energy production by mitochondria. OBJECTIVES: The authors aimed to determine the role of the calcineurin splicing variant CnAß1 in the context of cardiac hypertrophy and its mechanism of action. METHODS: Transgenic mice overexpressing CnAß1 specifically in cardiomyocytes and mice lacking the unique C-terminal domain in CnAß1 (CnAß1Δi12 mice) were used. Pressure overload hypertrophy was induced by transaortic constriction. Cardiac function was measured by echocardiography. Mice were characterized using various molecular analyses. RESULTS: In contrast to other calcineurin isoforms, the authors show here that cardiac-specific overexpression of CnAß1 in transgenic mice reduces cardiac hypertrophy and improves cardiac function. This effect is mediated by activation of serine and one-carbon metabolism, and the production of antioxidant mediators that prevent mitochondrial protein oxidation and preserve ATP production. The induction of enzymes involved in this metabolic pathway by CnAß1 is dependent on mTOR activity. Inhibition of serine and one-carbon metabolism blocks the beneficial effects of CnAß1. CnAß1Δi12 mice show increased cardiac hypertrophy and declined contractility. CONCLUSIONS: The metabolic reprogramming induced by CnAß1 redefines the role of calcineurin in the heart and shows for the first time that activation of the serine and one-carbon pathway has beneficial effects on cardiac hypertrophy and function, paving the way for new therapeutic approaches.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/metabolism , One-Carbon Group Transferases/metabolism , Serine/metabolism , Ventricular Function/drug effects , Animals , Calcineurin/pharmacology , Calcineurin/therapeutic use , Cardiomegaly/drug therapy , Humans , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Ventricular Function/physiology
13.
Cardiovasc Res ; 113(10): 1113-1123, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28472392

ABSTRACT

AIMS: Heart failure (HF) has become an epidemic and constitutes a major medical, social, and economic problem worldwide. Despite advances in medical treatment, HF prognosis remains poor. The development of efficient therapies is hampered by the lack of appropriate animal models in which HF can be reliably determined, particularly in mice. The development of HF in mice is often assumed based on the presence of cardiac dysfunction, but HF itself is seldom proved. Lung ultrasound (LUS) has become a helpful tool for lung congestion assessment in patients at all stages of HF. We aimed to apply this non-invasive imaging tool to evaluate HF in mouse models of both systolic and diastolic dysfunction. METHODS AND RESULTS: We used LUS to study HF in a mouse model of systolic dysfunction, dilated cardiomyopathy, and in a mouse model of diastolic dysfunction, diabetic cardiomyopathy. LUS proved to be a reliable and reproducible tool to detect pulmonary congestion in mice. The combination of LUS and echocardiography allowed discriminating those mice that develop HF from those that do not, even in the presence of evident cardiac dysfunction. The study showed that LUS can be used to identify the onset of HF decompensation and to evaluate the efficacy of therapies for this syndrome. CONCLUSIONS: This novel approach in mouse models of cardiac disease enables for the first time to adequately diagnose HF non-invasively in mice with preserved or reduced ejection fraction, and will pave the way to a better understanding of HF and to the development of new therapeutic approaches.


Subject(s)
Cardiomyopathy, Dilated/diagnostic imaging , Diabetic Cardiomyopathies/diagnostic imaging , Heart Failure/diagnostic imaging , Lung/diagnostic imaging , Pleural Effusion/diagnostic imaging , Pulmonary Edema/diagnostic imaging , Translational Research, Biomedical/methods , Ultrasonography/methods , Ventricular Function, Left , Animals , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/physiopathology , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/physiopathology , Diastole , Disease Models, Animal , Echocardiography, Doppler, Pulsed , Heart Failure/etiology , Heart Failure/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Pleural Effusion/etiology , Pleural Effusion/physiopathology , Predictive Value of Tests , Pulmonary Edema/etiology , Pulmonary Edema/physiopathology , Reproducibility of Results , Stroke Volume , Systole , Ventricular Function, Right
14.
Cell Chem Biol ; 23(11): 1372-1382, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27746127

ABSTRACT

Embryonic stem cells (ESC) have the potential to generate all the cell lineages that form the body. However, the molecular mechanisms underlying ESC differentiation and especially the role of alternative splicing in this process remain poorly understood. Here, we show that the alternative splicing regulator MBNL1 promotes generation of the atypical calcineurin Aß variant CnAß1 in mouse ESCs (mESC). CnAß1 has a unique C-terminal domain that drives its localization mainly to the Golgi apparatus by interacting with Cog8. CnAß1 regulates the intracellular localization and activation of the mTORC2 complex. CnAß1 knockdown results in delocalization of mTORC2 from the membrane to the cytoplasm, inactivation of the AKT/GSK3ß/ß-catenin signaling pathway, and defective mesoderm specification. In summary, here we unveil the structural basis for the mechanism of action of CnAß1 and its role in the differentiation of mESCs to the mesodermal lineage.


Subject(s)
Calcineurin/metabolism , Mouse Embryonic Stem Cells/cytology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Calcineurin/analysis , Cell Differentiation , Cell Line , Golgi Apparatus/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Mouse Embryonic Stem Cells/metabolism , Multiprotein Complexes/analysis , Signal Transduction , TOR Serine-Threonine Kinases/analysis
15.
Circ Cardiovasc Genet ; 8(5): 643-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26175529

ABSTRACT

BACKGROUND: Mutations in sarcomeric and cytoskeletal proteins are a major cause of hereditary cardiomyopathies, but our knowledge remains incomplete as to how the genetic defects execute their effects. METHODS AND RESULTS: We used cysteine and glycine-rich protein 3, a known cardiomyopathy gene, in a yeast 2-hybrid screen and identified zinc-finger and BTB domain-containing protein 17 (ZBTB17) as a novel interacting partner. ZBTB17 is a transcription factor that contains the peak association signal (rs10927875) at the replicated 1p36 cardiomyopathy locus. ZBTB17 expression protected cardiac myocytes from apoptosis in vitro and in a mouse model with cardiac myocyte-specific deletion of Zbtb17, which develops cardiomyopathy and fibrosis after biomechanical stress. ZBTB17 also regulated cardiac myocyte hypertrophy in vitro and in vivo in a calcineurin-dependent manner. CONCLUSIONS: We revealed new functions for ZBTB17 in the heart, a transcription factor that may play a role as a novel cardiomyopathy gene.


Subject(s)
Cardiomyopathies/genetics , Heart Failure/genetics , Nuclear Proteins/genetics , Animals , DNA-Binding Proteins , Heart/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nuclear Proteins/physiology , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/physiology , Rats , Stress, Physiological , Tissue Culture Techniques , Ubiquitin-Protein Ligases
16.
Cardiovasc Res ; 102(3): 396-406, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24667850

ABSTRACT

AIMS: Ventricular remodelling following myocardial infarction progressively leads to loss of contractile capacity and heart failure. Although calcineurin promotes maladaptive cardiac hypertrophy, we recently showed that the calcineurin splicing variant, CnAß1, has beneficial effects on the infarcted heart. However, whether this variant limits necrosis or improves remodelling is still unknown, precluding translation to the clinical arena. Here, we explored the effects and therapeutic potential of CnAß1 overexpression post-infarction. METHODS AND RESULTS: Double transgenic mice with inducible cardiomyocyte-specific overexpression of CnAß1 underwent left coronary artery ligation followed by reperfusion. Echocardiographic analysis showed depressed cardiac function in all infarcted mice 3 days post-infarction. Induction of CnAß1 overexpression 1 week after infarction improved function and reduced ventricular dilatation. CnAß1-overexpressing mice showed shorter, thicker scars, and reduced infarct expansion, accompanied by reduced myocardial remodelling. CnAß1 induced vascular endothelial growth factor (VEGF) expression in cardiomyocytes, which resulted in increased infarct vascularization. This paracrine angiogenic effect of CnAß1 was mediated by activation of the Akt/mammalian target of rapamycin pathway and VEGF. CONCLUSIONS: Our results indicate that CnAß1 exerts beneficial effects on the infarcted heart by promoting infarct vascularization and preventing infarct expansion. These findings emphasize the translational potential of CnAß1 for gene-based therapies.


Subject(s)
Calcineurin/physiology , Genetic Therapy , Myocardial Infarction/therapy , Ventricular Remodeling , Animals , Calcineurin/genetics , Mice , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/prevention & control , Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt/physiology , RNA Splicing , Signal Transduction/physiology
17.
Circulation ; 123(24): 2838-47, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21632490

ABSTRACT

BACKGROUND: Calcineurin is a calcium-regulated phosphatase that plays a major role in cardiac hypertrophy. We previously described that alternative splicing of the calcineurin Aß (CnAß) gene generates the CnAß1 isoform, with a unique C-terminal region that is different from the autoinhibitory domain present in all other CnA isoforms. In skeletal muscle, CnAß1 is necessary for myoblast proliferation and stimulates regeneration, reducing fibrosis and accelerating the resolution of inflammation. Its role in the heart is currently unknown. METHODS AND RESULTS: We generated transgenic mice overexpressing CnAß1 in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. In contrast to previous studies using an artificially truncated calcineurin, CnAß1 overexpression did not induce cardiac hypertrophy. Moreover, transgenic mice showed improved cardiac function and reduced scar formation after myocardial infarction, with reduced neutrophil and macrophage infiltration and decreased expression of proinflammatory cytokines. Immunoprecipitation and Western blot analysis showed interaction of CnAß1 with the mTOR complex 2 and activation of the Akt/SGK cardioprotective pathway in a PI3K-independent manner. In addition, gene expression profiling revealed that CnAß1 activated the transcription factor ATF4 downstream of the Akt/mTOR pathway to promote the amino acid biosynthesis program, to reduce protein catabolism, and to induce the antifibrotic and antiinflammatory factor growth differentiation factor 15, which protects the heart through Akt activation. CONCLUSIONS: Calcineurin Aß1 shows a unique mode of action that improves cardiac function after myocardial infarction, activating different cardioprotective pathways without inducing maladaptive hypertrophy. These features make CnAß1 an attractive candidate for the development of future therapeutic approaches.


Subject(s)
Calcineurin/genetics , Heart/physiology , Myocardial Contraction/physiology , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Calcineurin/metabolism , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Fibrosis , Gene Expression Profiling , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocarditis/genetics , Myocarditis/metabolism , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Isoforms/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...