Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Nucleic Acids Res ; 52(8): 4215-4233, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38364861

ABSTRACT

The limited regenerative capacity of the human heart contributes to high morbidity and mortality worldwide. In contrast, zebrafish exhibit robust regenerative capacity, providing a powerful model for studying how to overcome intrinsic epigenetic barriers maintaining cardiac homeostasis and initiate regeneration. Here, we present a comprehensive analysis of the histone modifications H3K4me1, H3K4me3, H3K27me3 and H3K27ac during various stages of zebrafish heart regeneration. We found a vast gain of repressive chromatin marks one day after myocardial injury, followed by the acquisition of active chromatin characteristics on day four and a transition to a repressive state on day 14, and identified distinct transcription factor ensembles associated with these events. The rapid transcriptional response involves the engagement of super-enhancers at genes implicated in extracellular matrix reorganization and TOR signaling, while H3K4me3 breadth highly correlates with transcriptional activity and dynamic changes at genes involved in proteolysis, cell cycle activity, and cell differentiation. Using loss- and gain-of-function approaches, we identified transcription factors in cardiomyocytes and endothelial cells influencing cardiomyocyte dedifferentiation or proliferation. Finally, we detected significant evolutionary conservation between regulatory regions that drive zebrafish and neonatal mouse heart regeneration, suggesting that reactivating transcriptional and epigenetic networks converging on these regulatory elements might unlock the regenerative potential of adult human hearts.


Subject(s)
Chromatin , Gene Regulatory Networks , Heart , Animals , Humans , Mice , Cell Differentiation , Chromatin/metabolism , Chromatin/genetics , Epigenesis, Genetic , Histone Code , Histones/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Regeneration/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish/genetics
2.
Circ Res ; 133(10): 842-857, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37800327

ABSTRACT

BACKGROUND: Advanced age is unequivocally linked to the development of cardiovascular disease; however, the mechanisms resulting in reduced endothelial cell regeneration remain poorly understood. Here, we investigated novel mechanisms involved in endothelial cell senescence that impact endothelial cell transcription and vascular repair after injury. METHODS: Native endothelial cells were isolated from young (20±3.4 years) and aged (80±2.3 years) individuals and subjected to molecular analyses to assess global transcriptional and metabolic changes. In vitro studies were conducted using primary human and murine endothelial cells. A murine aortic re-endothelialization model was used to examine endothelial cell regenerative capacity in vivo. RESULTS: RNA sequencing of native endothelial cells revealed that aging resulted in p53-mediated reprogramming to express senescence-associated genes and suppress glycolysis. Reduced glucose uptake and ATP contributed to attenuated assembly of the telomerase complex, which was required for endothelial cell proliferation. Enhanced p53 activity in aging was linked to its acetylation on K120 due to enhanced activity of the acetyltransferase MOZ (monocytic leukemic zinc finger). Mechanistically, p53 acetylation and translocation were, at least partially, attributed to the loss of the vasoprotective enzyme, CSE (cystathionine γ-lyase). CSE physically anchored p53 in the cytosol to prevent its nuclear translocation and CSE absence inhibited AKT (Protein kinase B)-mediated MOZ phosphorylation, which in turn increased MOZ activity and subsequently p53 acetylation. In mice, the endothelial cell-specific deletion of CSE activated p53, induced premature endothelial senescence, and arrested vascular repair after injury. In contrast, the adeno-associated virus 9-mediated re-expression of an active CSE mutant retained p53 in the cytosol, maintained endothelial glucose metabolism and proliferation, and prevented endothelial cell senescence. Adenoviral overexpression of CSE in native endothelial cells from aged individuals maintained low p53 activity and reactivated telomerase to revert endothelial cell senescence. CONCLUSIONS: Aging-associated impairment of vascular repair is partly determined by the vasoprotective enzyme CSE.


Subject(s)
Hydrogen Sulfide , Telomerase , Animals , Humans , Mice , Cellular Senescence , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Endothelial Cells/metabolism , Hydrogen Sulfide/metabolism , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Angiogenesis ; 27(2): 193-209, 2024 May.
Article in English | MEDLINE | ID: mdl-38070064

ABSTRACT

Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor ß and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.


Subject(s)
Endothelial Cells , Vascular Diseases , Humans , Proto-Oncogene Proteins c-sis/metabolism , Endothelial Cells/metabolism , Vascular Diseases/metabolism , Capillaries/metabolism , Pericytes/metabolism
4.
Nat Methods ; 18(9): 1068-1074, 2021 09.
Article in English | MEDLINE | ID: mdl-34480152

ABSTRACT

In general, mRNAs are assumed to be loaded with ribosomes instantly upon entry into the cytoplasm. To measure ribosome density (RD) on nascent mRNA, we developed nascent Ribo-Seq by combining Ribo-Seq with progressive 4-thiouridine labeling. In mouse macrophages, we determined experimentally the lag between the appearance of nascent mRNA and its association with ribosomes, which was calculated to be 20-22 min for bulk mRNA. In mouse embryonic stem cells, nRibo-Seq revealed an even stronger lag of 35-38 min in ribosome loading. After stimulation of macrophages with lipopolysaccharide, the lag between cytoplasmic and translated mRNA leads to uncoupling between input and ribosome-protected fragments, which gives rise to distorted RD measurements under conditions where mRNA amounts are far from steady-state expression. As a result, we demonstrate that transcriptional changes affect RD in a passive way.


Subject(s)
Protein Biosynthesis , Ribosomes/genetics , Ribosomes/metabolism , Sequence Analysis, RNA/methods , Animals , Cytoplasm/genetics , Kinetics , Lipopolysaccharides/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/physiology , RAW 264.7 Cells , RNA, Messenger/genetics , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/genetics , Ribosomes/drug effects , Time Factors
5.
Arterioscler Thromb Vasc Biol ; 43(10): 1967-1989, 2023 10.
Article in English | MEDLINE | ID: mdl-37650327

ABSTRACT

BACKGROUND: Endothelial cells (ECs) are primed to respond to various signaling cues. For example, TGF (transforming growth factor)-ß has major effects on EC function and phenotype by driving ECs towards a more mesenchymal state (ie, triggering endothelial to mesenchymal activation), a dynamic process associated with cardiovascular diseases. Although transcriptional regulation triggered by TGF-ß in ECs is well characterized, post-transcriptional regulatory mechanisms induced by TGF-ß remain largely unknown. METHODS: Using RNA interactome capture, we identified global TGF-ß driven changes in RNA-binding proteins in ECs. We investigated specific changes in the RNA-binding patterns of hnRNP H1 (heterogeneous nuclear ribonucleoprotein H1) and Csde1 (cold shock domain containing E1) using RNA immunoprecipitation and overlapped this with RNA-sequencing data after knockdown of either protein for functional insight. Using a modified proximity ligation assay, we visualized the specific interactions between hnRNP H1 and Csde1 and target RNAs in situ both in vitro and in mouse heart sections. RESULTS: Characterization of TGF-ß-regulated RBPs (RNA-binding proteins) revealed hnRNP H1 and Csde1 as key regulators of the cellular response to TGF-ß at the post-transcriptional level, with loss of either protein-promoting mesenchymal activation in ECs. We found that TGF-ß drives an increase in binding of hnRNP H1 to its target RNAs, offsetting mesenchymal activation, but a decrease in Csde1 RNA-binding, facilitating this process. Both, hnRNP H1 and Csde1, dynamically bind and regulate specific subsets of mRNAs related to mesenchymal activation and endothelial function. CONCLUSIONS: Together, we show that RBPs play a key role in the endothelial response to TGF-ß stimulation at the post-transcriptional level and that the RBPs hnRNP H1 and Csde1 serve to maintain EC function and counteract mesenchymal activation. We propose that TGF-ß profoundly modifies RNA-protein interaction entailing feedback and feed-forward control at the post-transcriptional level, to fine-tune mesenchymal activation in ECs.


Subject(s)
Endothelial Cells , Transforming Growth Factor beta , Mice , Animals , Transforming Growth Factor beta/metabolism , Endothelial Cells/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA
6.
Adv Exp Med Biol ; 1441: 271-294, 2024.
Article in English | MEDLINE | ID: mdl-38884717

ABSTRACT

Cardiovascular diseases, both congenital and acquired, are the leading cause of death worldwide, associated with significant health consequences and economic burden. Due to major advances in surgical procedures, most patients with congenital heart disease (CHD) survive into adulthood but suffer from previously unrecognized long-term consequences, such as early-onset heart failure. Therefore, understanding the molecular mechanisms resulting in heart defects and the lifelong complications due to hemodynamic overload are of utmost importance. Congenital heart disease arises in the first trimester of pregnancy, due to defects in the complex morphogenetic patterning of the heart. This process is coordinated through a complicated web of intercellular communication between the epicardium, the endocardium, and the myocardium. In the postnatal heart, similar crosstalk between cardiomyocytes, endothelial cells, and fibroblasts exists during pathological hemodynamic overload that emerges as a consequence of a congenital heart defect. Ultimately, communication between cells triggers the activation of intracellular signaling circuits, which allow fine coordination of cardiac development and function. Here, we review the inter- and intracellular signaling mechanisms in the heart as they were discovered mainly in genetically modified mice.


Subject(s)
Cell Communication , Heart Defects, Congenital , Signal Transduction , Humans , Animals , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocardium/metabolism , Myocardium/pathology , Mice , Pregnancy , Heart/embryology , Heart/growth & development
7.
Basic Res Cardiol ; 117(1): 15, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35286475

ABSTRACT

Hyperactivity of the sympathetic nervous system is a major driver of cardiac remodeling, exerting its effects through both α-, and ß-adrenoceptors (α-, ß-ARs). As the relative contribution of subtype α1-AR to cardiac stress responses remains poorly investigated, we subjected mice to either subcutaneous perfusion with the ß-AR agonist isoprenaline (ISO, 30 mg/kg × day) or to a combination of ISO and the stable α1-AR agonist phenylephrine (ISO/PE, 30 mg/kg × day each). Telemetry analysis revealed similar hemodynamic responses under both ISO and ISO/PE treatment i.e., permanently increased heart rates and only transient decreases in mean blood pressure during the first 24 h. Echocardiography and single cell analysis after 1 week of exposure showed that ISO/PE-, but not ISO-treated animals established α1-AR-mediated inotropic responsiveness to acute adrenergic stimulation. Morphologically, additional PE perfusion limited concentric cardiomyocyte growth and enhanced cardiac collagen deposition during 7 days of treatment. Time-course analysis demonstrated a diverging development in transcriptional patterns at day 4 of treatment i.e., increased expression of selected marker genes Xirp2, Nppa, Tgfb1, Col1a1, Postn under chronic ISO/PE treatment which was either less pronounced or absent in the ISO group. Transcriptome analyses at day 4 via RNA sequencing demonstrated that additional PE treatment caused a marked upregulation of genes allocated to extracellular matrix and fiber organization along with a more pronounced downregulation of genes involved in metabolic processes, muscle adaptation and cardiac electrophysiology. Consistently, transcriptome changes under ISO/PE challenge more effectively recapitulated early transcriptional alterations in pressure overload-induced experimental heart failure and in human hypertrophic cardiomyopathy.


Subject(s)
Heart , Receptors, Adrenergic, alpha-1 , Animals , Isoproterenol/pharmacology , Mice , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta
8.
J Hepatol ; 74(2): 380-393, 2021 02.
Article in English | MEDLINE | ID: mdl-32916216

ABSTRACT

BACKGROUND & AIMS: Angiocrine signaling by liver sinusoidal endothelial cells (LSECs) regulates hepatic functions such as growth, metabolic maturation, and regeneration. Recently, we identified GATA4 as the master regulator of LSEC specification during development. Herein, we studied the role of endothelial GATA4 in the adult liver and in hepatic pathogenesis. METHODS: We generated adult Clec4g-icretg/0xGata4fl/fl (Gata4LSEC-KO) mice with LSEC-specific depletion of Gata4. Livers were analyzed by histology, electron microscopy, immunohistochemistry/immunofluorescence, in situ hybridization, and LSECs were isolated for gene expression profiling, ChIP- and ATAC-sequencing. Partial hepatectomy was performed to assess regeneration. We used choline-deficient, l-amino acid-defined (CDAA) diet and chronic carbon tetrachloride exposure to model liver fibrosis. Human single cell RNA-seq data sets were analyzed for endothelial alterations in healthy and cirrhotic livers. RESULTS: Genetic Gata4 deficiency in LSECs of adult mice caused perisinusoidal liver fibrosis, hepatopathy and impaired liver regeneration. Sinusoidal capillarization and LSEC-to-continuous endothelial transdifferentiation were accompanied by a profibrotic angiocrine switch involving de novo endothelial expression of hepatic stellate cell-activating cytokine PDGFB. Increased chromatin accessibility and amplification by activated MYC mediated angiocrine Pdgfb expression. As observed in Gata4LSEC-KO livers, CDAA diet-induced perisinusoidal liver fibrosis was associated with GATA4 repression, MYC activation and a profibrotic angiocrine switch in LSECs. Comparison of CDAA-fed Gata4LSEC-KO and control mice demonstrated that endothelial GATA4 indeed protects against dietary-induced perisinusoidal liver fibrosis. In human cirrhotic livers, GATA4-positive LSECs and endothelial GATA4 target genes were reduced, while non-LSEC endothelial cells and MYC target genes including PDGFB were enriched. CONCLUSIONS: Endothelial GATA4 protects against perisinusoidal liver fibrosis by repressing MYC activation and profibrotic angiocrine signaling at the chromatin level. Therapies targeting the GATA4/MYC/PDGFB/PDGFRß axis offer a promising strategy for prevention and treatment of liver fibrosis. LAY SUMMARY: The liver vasculature is supposed to play a major role in the development of liver fibrosis and cirrhosis, which can lead to liver failure and liver cancer. Herein, we discovered that structural and transcriptional changes induced by genetic deletion of the transcription factor GATA4 in the hepatic endothelium were sufficient to cause liver fibrosis. Activation of the transcription factor MYC and de novo expression of the "angiocrine" growth factor PDGFB were identified as downstream drivers of fibrosis and as potential therapeutic targets for this potentially fatal disease.


Subject(s)
Endothelial Cells/metabolism , GATA4 Transcription Factor/metabolism , Liver Cirrhosis , Liver , Lymphokines , Platelet-Derived Growth Factor , Animals , Chromatin/metabolism , Drug Discovery , Gene Expression Profiling , Hepatic Stellate Cells/metabolism , Humans , Liver/blood supply , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , Liver Regeneration/physiology , Lymphokines/genetics , Lymphokines/metabolism , Mice , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Zinc Fingers
10.
Basic Res Cardiol ; 116(1): 26, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33876316

ABSTRACT

Heart failure due to high blood pressure or ischemic injury remains a major problem for millions of patients worldwide. Despite enormous advances in deciphering the molecular mechanisms underlying heart failure progression, the cell-type specific adaptations and especially intercellular signaling remain poorly understood. Cardiac fibroblasts express high levels of cardiogenic transcription factors such as GATA-4 and GATA-6, but their role in fibroblasts during stress is not known. Here, we show that fibroblast GATA-4 and GATA-6 promote adaptive remodeling in pressure overload induced cardiac hypertrophy. Using a mouse model with specific single or double deletion of Gata4 and Gata6 in stress activated fibroblasts, we found a reduced myocardial capillarization in mice with Gata4/6 double deletion following pressure overload, while single deletion of Gata4 or Gata6 had no effect. Importantly, we confirmed the reduced angiogenic response using an in vitro co-culture system with Gata4/6 deleted cardiac fibroblasts and endothelial cells. A comprehensive RNA-sequencing analysis revealed an upregulation of anti-angiogenic genes upon Gata4/6 deletion in fibroblasts, and siRNA mediated downregulation of these genes restored endothelial cell growth. In conclusion, we identified a novel role for the cardiogenic transcription factors GATA-4 and GATA-6 in heart fibroblasts, where both proteins act in concert to promote myocardial capillarization and heart function by directing intercellular crosstalk.


Subject(s)
Cardiomegaly/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Ventricular Remodeling , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Aorta/physiopathology , Aorta/surgery , Arterial Pressure , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cell Communication , Cells, Cultured , Constriction , Disease Models, Animal , Fibroblasts/pathology , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Mice, Knockout , Microvascular Density , Myocardium/pathology , Signal Transduction
11.
Cytokine ; 133: 155142, 2020 09.
Article in English | MEDLINE | ID: mdl-32485621

ABSTRACT

BACKGROUND: Pathophysiology of aneurysmal subarachnoid hemorrhage (aSAH) is highly complex. Bleeding from ruptured aneurysm causes increase in intracranial pressure that disrupts blood-brain barrier leading to infiltration of peripheral immune cells. Interactions between the infiltrated leukocytes and the resident brain cells in the injured tissue mainly determine the delayed tissue damage. Recruitment of leukocytes in the injured brain is mainly mediated by the chemokines. Chemokine C-C motif ligand 5 (CCL5) is a potent pro-inflammatory chemokine shown to be upregulated in preclinical SAH studies. However, detailed clinical investigations exploring the association of cerebrospinal fluid (CSF) and systemic CCL5 and post-aSAH complications and clinical outcome are still lacking. This study investigated CSF and systemic CCL5 after aSAH and its association with clinical outcome and post-aSAH complications. METHODS: CSF and serum from control and aSAH patients were obtained after centrifugation of the CSF and peripheral blood, and were preserved at -80 °C until quantification by an enzyme-linked immunoassay. Patient pertinent data, post-aSAH complications and clinical outcome (modified Rankin scale [mRS] and Glasgow outcome scale [GOS]) were retrieved from patient records. RESULTS: A significant increase in CSF and serum CCL5 levels was observed on post-aSAH day 1 and day 7 compared to control patients. Dichotomization of patients to poor (mRS 3-6 or GOS 1-3) and good (mRS 0-2 or GOS 4-5) clinical outcomes showed significantly higher serum CCL5 levels in patients with good clinical outcome at discharge, but lower CSF CCL5 levels. Interestingly, significantly lower serum CCL5 levels were observed on post-aSAH day 7 in patients who have additional intracerebral bleeding or the patients who developed chronic hydrocephalus or pneumonia. Whereas, CSF CCL5 levels significantly increased on post-aSAH day 1 in patients developing chronic hydrocephalus, delayed ischemic neurological deficits and intraventricular hemorrhage. CSF CCL5 levels on post-aSAH day 1 were correlated with poor clinical outcome, however, serum CCL5 levels on post-aSAH day 7 were correlated with good clinical outcome. CONCLUSION: Systemic and CSF CCL5 levels were elevated after aSAH and levels of serum CCL5 on day 7 were associated independently with clinical outcome (GOS and mRS) at discharge. Therapeutic approaches targeting CCL5 might be beneficial in aSAH.


Subject(s)
Biomarkers/metabolism , Cerebrospinal Fluid/metabolism , Chemokine CCL5/metabolism , Subarachnoid Hemorrhage/metabolism , Female , Humans , Male , Middle Aged , Prospective Studies , Up-Regulation/physiology
12.
Stem Cells ; 33(4): 1113-29, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524439

ABSTRACT

During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.


Subject(s)
Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Animals , Cell Lineage/physiology , Embryonic Stem Cells/metabolism , HEK293 Cells , Homeobox Protein Nkx-2.5 , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding/physiology , Xenopus
13.
Cereb Cortex ; 25(10): 3406-19, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25037921

ABSTRACT

The chromatin-remodeling protein Satb2 plays a role in the generation of distinct subtypes of neocortical pyramidal neurons. Previous studies have shown that Satb2 is required for normal development of callosal projection neurons (CPNs), which fail to extend axons callosally in the absence of Satb2 and instead project subcortically. Here we conditionally delete Satb2 from the developing neocortex and find that neurons in the upper layers adopt some electrophysiological properties characteristic of deep layer neurons, but projections from the superficial layers do not contribute to the aberrant subcortical projections seen in Satb2 mutants. Instead, axons from deep layer CPNs descend subcortically in the absence of Satb2. These data demonstrate distinct developmental roles of Satb2 in regulating the fates of upper and deep layer neurons. Unexpectedly, Satb2 mutant brains also display changes in gene expression by subcerebral projection neurons (SCPNs), accompanied by a failure of corticospinal tract (CST) formation. Altering the timing of Satb2 ablation reveals that SCPNs require an early expression of Satb2 for differentiation and extension of the CST, suggesting that early transient expression of Satb2 in these cells plays an essential role in development. Collectively these data show that Satb2 is required by both CPNs and SCPNs for proper differentiation and axon pathfinding.


Subject(s)
Axons/physiology , Cell Differentiation , Cerebral Cortex/embryology , Corpus Callosum/embryology , Matrix Attachment Region Binding Proteins/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Axons/metabolism , Brain/embryology , Brain/metabolism , Cerebral Cortex/metabolism , Corpus Callosum/metabolism , Female , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/metabolism , Neurons/metabolism , Somatosensory Cortex/embryology , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Circ Res ; 113(7): 856-62, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23856522

ABSTRACT

RATIONALE: The developmental role of the H3K27 demethylases Jmjd3, especially its epigenetic regulation at target genes in response to upstream developmental signaling, is unclear. OBJECTIVE: To determine the role of Jmjd3 during mesoderm and cardiovascular lineage commitment. METHODS AND RESULTS: Ablation of Jmjd3 in mouse embryonic stem cells does not affect the maintenance of pluripotency and self-renewal but compromised mesoderm and subsequent endothelial and cardiac differentiation. Jmjd3 reduces H3K27me3 marks at the Brachyury promoter and facilitates the recruitment of ß-catenin, which is critical for Wnt signal-induced mesoderm differentiation. CONCLUSIONS: These data demonstrate that Jmjd3 is required for mesoderm differentiation and cardiovascular lineage commitment.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Endothelium, Vascular/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesoderm/cytology , Myocytes, Cardiac/cytology , Animals , Cell Line , Cell Lineage , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Fetal Proteins/genetics , Fetal Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Mesoderm/metabolism , Mice , Mutation , Promoter Regions, Genetic , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
15.
Proc Natl Acad Sci U S A ; 109(47): 19071-8, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23144223

ABSTRACT

Neurons within each layer in the mammalian cortex have stereotypic projections. Four genes-Fezf2, Ctip2, Tbr1, and Satb2-regulate these projection identities. These genes also interact with each other, and it is unclear how these interactions shape the final projection identity. Here we show, by generating double mutants of Fezf2, Ctip2, and Satb2, that cortical neurons deploy a complex genetic switch that uses mutual repression to produce subcortical or callosal projections. We discovered that Tbr1, EphA4, and Unc5H3 are critical downstream targets of Satb2 in callosal fate specification. This represents a unique role for Tbr1, implicated previously in specifying corticothalamic projections. We further show that Tbr1 expression is dually regulated by Satb2 and Ctip2 in layers 2-5. Finally, we show that Satb2 and Fezf2 regulate two disease-related genes, Auts2 (Autistic Susceptibility Gene2) and Bhlhb5 (mutated in Hereditary Spastic Paraplegia), providing a molecular handle to investigate circuit disorders in neurodevelopmental diseases.


Subject(s)
Gene Regulatory Networks , Neocortex/growth & development , Neocortex/metabolism , Neurons/metabolism , Repressor Proteins/metabolism , Alkaline Phosphatase/metabolism , Animals , Axons/enzymology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Isoenzymes/metabolism , Mice , Mutation/genetics , Nerve Tissue Proteins/metabolism , Netrin Receptors , Nuclear Proteins/metabolism , Protein Binding , Receptor, EphA4/metabolism , Receptors, Cell Surface/metabolism , Repressor Proteins/genetics , T-Box Domain Proteins , Thalamus/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism
16.
Gene ; 893: 147915, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-37866662

ABSTRACT

S-adenosylmethionine (SAM) represents a potent inhibitor of cancer cell proliferation, migration, and invasionin vitro.The underlying mechanisms remain elusive. Here, we examined, if treatment with SAM may cause alterations in the methylation of the histone marks H3K4me3 and H3K27me3, which are both known to play important roles in the initiation and progression of prostate cancer. We treated PC-3 cells with 200 µmol SAM, a concentration known to cause anticancerogenic effects, followed by ChIP-sequencing for H3K4me3 and H3K27me3. We detected 236 differentially methylated regions for H3K27me3 and 560 differentially methylated regions for H3K4me3. GO Term enrichment showed upregulation of anticancerogenic, as well as downregulation of cancerogenic related biological processes, molecular functions, and pathways. Furthermore, we compared specific methylation profiles of SAM treated samples to gene expression changes (RNA-Seq). 35 upregulated and 56 downregulated genes (total: 604 differentially expressed genes) could be related to hypomethylated and hypermethylated regions. 17 upregulated genes could be identified as tumor suppressor genes, 45 downregulated genes in contrast are considered as oncogenes. As a conclusion it can be stated that SAM treatment of prostate cancer cells resulted in alterations of H3K4me3 and H3K27me3 methylation profiles. Gene to peak annotation, alignment with results of a transcriptome study as well as GO-term analysis underpinned the biological relevance of methylation changes.


Subject(s)
Histones , Prostatic Neoplasms , Male , Humans , Methylation , Histones/metabolism , S-Adenosylmethionine/pharmacology , S-Adenosylmethionine/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , DNA Methylation
17.
Sci Transl Med ; 16(736): eabq4581, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38416842

ABSTRACT

Fibrosis is a hallmark of chronic disease. Although fibroblasts are involved, it is unclear to what extent endothelial cells also might contribute. We detected increased expression of the transcription factor Sox9 in endothelial cells in several different mouse fibrosis models. These models included systolic heart failure induced by pressure overload, diastolic heart failure induced by high-fat diet and nitric oxide synthase inhibition, pulmonary fibrosis induced by bleomycin treatment, and liver fibrosis due to a choline-deficient diet. We also observed up-regulation of endothelial SOX9 in cardiac tissue from patients with heart failure. To test whether SOX9 induction was sufficient to cause disease, we generated mice with endothelial cell-specific overexpression of Sox9, which promoted fibrosis in multiple organs and resulted in signs of heart failure. Endothelial Sox9 deletion prevented fibrosis and organ dysfunction in the two mouse models of heart failure as well as in the lung and liver fibrosis mouse models. Bulk and single-cell RNA sequencing of mouse endothelial cells across multiple vascular beds revealed that SOX9 induced extracellular matrix, growth factor, and inflammatory gene expression, leading to matrix deposition by endothelial cells. Moreover, mouse endothelial cells activated neighboring fibroblasts that then migrated and deposited matrix in response to SOX9, a process partly mediated by the secreted growth factor CCN2, a direct SOX9 target; endothelial cell-specific Sox9 deletion reversed these changes. These findings suggest a role for endothelial SOX9 as a fibrosis-promoting factor in different mouse organs during disease and imply that endothelial cells are an important regulator of fibrosis.


Subject(s)
Heart Failure , Transcription Factors , Animals , Humans , Mice , Disease Models, Animal , Endothelial Cells , Fibrosis , Intercellular Signaling Peptides and Proteins , Liver Cirrhosis/complications , SOX9 Transcription Factor/genetics
18.
Cells Dev ; 175: 203857, 2023 09.
Article in English | MEDLINE | ID: mdl-37257755

ABSTRACT

The heart is a complex organ composed of distinct cell types, such as cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, neuronal cells and immune cells. All these cell types contribute to the structural, electrical and mechanical properties of the heart. Genetic manipulation and lineage tracing studies in mice have been instrumental in gaining critical insights into the networks regulating cardiac cell lineage specification, cell fate and plasticity. Such knowledge has been of fundamental importance for the development of efficient protocols for the directed differentiation of pluripotent stem cells (PSCs) in highly specialized cardiac cell types. In this review, we summarize the evolution and current advances in protocols for cardiac subtype specification, maturation, and assembly in cardiac microtissues and organoids.


Subject(s)
Endothelial Cells , Pluripotent Stem Cells , Humans , Mice , Animals , Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Cell Differentiation/genetics , Fibroblasts
19.
Cells ; 12(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36899919

ABSTRACT

Mutations in the gene for lamin A/C (LMNA) cause a diverse range of diseases known as laminopathies. LMNA-related cardiomyopathy is a common inherited heart disease and is highly penetrant with a poor prognosis. In the past years, numerous investigations using mouse models, stem cell technologies, and patient samples have characterized the phenotypic diversity caused by specific LMNA variants and contributed to understanding the molecular mechanisms underlying the pathogenesis of heart disease. As a component of the nuclear envelope, LMNA regulates nuclear mechanostability and function, chromatin organization, and gene transcription. This review will focus on the different cardiomyopathies caused by LMNA mutations, address the role of LMNA in chromatin organization and gene regulation, and discuss how these processes go awry in heart disease.


Subject(s)
Cardiomyopathies , Lamin Type A , Animals , Mice , Cardiomyopathies/genetics , Chromatin , Epigenesis, Genetic , Heart , Lamin Type A/metabolism , Humans
20.
J Clin Invest ; 133(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37490341

ABSTRACT

Vascular networks form, remodel, and mature under the influence of both fluid shear stress (FSS) and soluble factors. Physiological FSS promotes and maintains vascular stability via synergy with bone morphogenic proteins 9 and 10 (BMP9 and BMP10). Conversely, mutation of the BMP receptors activin-like kinase 1 (ALK1), endoglin (ENG), or the downstream effector, SMAD family member 4 (SMAD4) leads to hereditary hemorrhagic telangiectasia (HHT), characterized by fragile and leaky arterial-venous malformations (AVMs). How endothelial cells (ECs) integrate FSS and BMP signals in vascular development and homeostasis and how mutations give rise to vascular malformations is not well understood. Here, we aimed to elucidate the mechanism of synergy between FSS and SMAD signaling in vascular stability and how disruption of this synergy leads to AVMs. We found that loss of Smad4 increased the sensitivity of ECs to flow by lowering the FSS set point, with resulting AVMs exhibiting features of excessive flow-mediated morphological responses. Mechanistically, loss of SMAD4 disinhibits flow-mediated KLF4-TIE2-PI3K/Akt signaling, leading to cell cycle progression-mediated loss of arterial identity due to KLF4-mediated repression of cyclin dependent Kinase (CDK) inhibitors CDKN2A and CDKN2B. Thus, AVMs caused by Smad4 deletion are characterized by chronic high flow remodeling with excessive EC proliferation and loss of arterial identity as triggering events.


Subject(s)
Arteriovenous Malformations , Telangiectasia, Hereditary Hemorrhagic , Mice , Animals , Arteriovenous Malformations/genetics , Arteriovenous Malformations/metabolism , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice, Knockout , Telangiectasia, Hereditary Hemorrhagic/genetics , Bone Morphogenetic Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL