Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 117(8): 1908-1922, 2021 07 07.
Article in English | MEDLINE | ID: mdl-32777030

ABSTRACT

AIMS: Arrhythmias and sudden cardiac death (SCD) occur commonly in patients with heart failure. We found T-box 5 (TBX5) dysregulated in ventricular myocardium from heart failure patients and thus we hypothesized that TBX5 reduction contributes to arrhythmia development in these patients. To understand the underlying mechanisms, we aimed to reveal the ventricular TBX5-dependent transcriptional network and further test the therapeutic potential of TBX5 level normalization in mice with documented arrhythmias. METHODS AND RESULTS: We used a mouse model of TBX5 conditional deletion in ventricular cardiomyocytes. Ventricular (v) TBX5 loss in mice resulted in mild cardiac dysfunction and arrhythmias and was associated with a high mortality rate (60%) due to SCD. Upon angiotensin stimulation, vTbx5KO mice showed exacerbated cardiac remodelling and dysfunction suggesting a cardioprotective role of TBX5. RNA-sequencing of a ventricular-specific TBX5KO mouse and TBX5 chromatin immunoprecipitation was used to dissect TBX5 transcriptional network in cardiac ventricular tissue. Overall, we identified 47 transcripts expressed under the control of TBX5, which may have contributed to the fatal arrhythmias in vTbx5KO mice. These included transcripts encoding for proteins implicated in cardiac conduction and contraction (Gja1, Kcnj5, Kcng2, Cacna1g, Chrm2), in cytoskeleton organization (Fstl4, Pdlim4, Emilin2, Cmya5), and cardiac protection upon stress (Fhl2, Gpr22, Fgf16). Interestingly, after TBX5 loss and arrhythmia development in vTbx5KO mice, TBX5 protein-level normalization by systemic adeno-associated-virus (AAV) 9 application, re-established TBX5-dependent transcriptome. Consequently, cardiac dysfunction was ameliorated and the propensity of arrhythmia occurrence was reduced. CONCLUSIONS: This study uncovers a novel cardioprotective role of TBX5 in the adult heart and provides preclinical evidence for the therapeutic value of TBX5 protein normalization in the control of arrhythmia.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Death, Sudden, Cardiac/prevention & control , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/therapy , T-Box Domain Proteins/metabolism , Ventricular Dysfunction, Left/therapy , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Chromatin Immunoprecipitation Sequencing , Death, Sudden, Cardiac/etiology , Disease Models, Animal , Gene Expression Profiling , Genetic Therapy , Heart Rate , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Isolated Heart Preparation , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , T-Box Domain Proteins/genetics , Transcription, Genetic , Transcriptome , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling
2.
Stem Cells ; 31(5): 928-40, 2013 May.
Article in English | MEDLINE | ID: mdl-23341242

ABSTRACT

The multiphasic regulation of the Wnt/ß-catenin canonical pathway is essential for cardiogenesis in vivo and in vitro. To achieve tight regulation of the Wnt/ß-catenin signaling, tissue- and cell-specific coactivators and repressors need to be recruited. The identification of such factors may help to elucidate mechanisms leading to enhanced cardiac differentiation efficiency in vitro as well as promote regeneration in vivo. Using a yeast-two-hybrid screen, we identified four-and-a-half-LIM-domain 2 (FHL2) as a cardiac-specific ß-catenin interaction partner and activator of Wnt/ß-catenin-dependent transcription. We analyzed the role of this interaction for early cardiogenesis in an in vitro model by making use of embryoid body cultures from mouse embryonic stem cells (ESCs). In this model, stable FHL2 gain-of-function promoted mesodermal cell formation and cell proliferation while arresting cardiac differentiation in an early cardiogenic mesodermal progenitor state. Mechanistically, FHL2 overexpression enhanced nuclear accumulation of ß-catenin and activated Wnt/ß-catenin-dependent transcription leading to sustained upregulation of the early cardiogenic gene Igfbp5. In an alternative P19 cell model, transient FHL2 overexpression led to early activation of Wnt/ß-catenin-dependent transcription, but not sustained high-level of Igfbp5 expression. This resulted in enhanced cardiogenesis. We propose that early Wnt/ß-catenin-dependent transcriptional activation mediated by FHL2 is important for the transition to and expansion of early cardiogenic mesodermal cells. Collectively, our findings offer mechanistic insight into the early cardiogenic code and may be further exploited to enhance cardiac progenitor cell activity in vitro and in vivo.


Subject(s)
Embryonic Stem Cells/physiology , LIM-Homeodomain Proteins/genetics , Muscle Proteins/genetics , Myocytes, Cardiac/cytology , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Muscle Proteins/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , Transfection , beta Catenin/biosynthesis , beta Catenin/metabolism
3.
EMBO Mol Med ; 4(9): 992-1007, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22767436

ABSTRACT

Wnt/ß-catenin signalling controls adult heart remodelling in part via regulation of cardiac progenitor cell (CPC) differentiation. An enhanced understanding of mechanisms controlling CPC biology might facilitate the development of new therapeutic strategies in heart failure. We identified and characterized a novel cardiac interaction between Krueppel-like factor 15 and components of the Wnt/ß-catenin pathway leading to inhibition of transcription. In vitro mutation, reporter assays and co-localization analyses revealed that KLF15 requires both the C-terminus, necessary for nuclear localization, and a minimal N-terminal regulatory region to inhibit transcription. In line with this, functional Klf15 knock-out mice exhibited cardiac ß-catenin transcriptional activation along with functional cardiac deterioration in normal homeostasis and upon hypertrophy. We further provide in vivo and in vitro evidences for preferential endothelial lineage differentiation of CPCs upon KLF15 deletion. Via inhibition of ß-catenin transcription, KLF15 controls CPC homeostasis in the adult heart similar to embryonic cardiogenesis. This knowledge may provide a tool for reactivation of this apparently dormant CPC population in the adult heart and thus be an attractive approach to enhance endogenous cardiac repair.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Heart/physiology , Stem Cells/physiology , Transcription Factors/metabolism , Wnt Proteins/biosynthesis , beta Catenin/biosynthesis , Animals , Down-Regulation , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Transcription, Genetic , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors
4.
Biochim Biophys Acta ; 1811(6): 361-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21420506

ABSTRACT

Pancreatic ß-cells have a deficit of scavenging enzymes such as catalase (Cat) and glutathione peroxidase (GPx) and therefore are susceptible to oxidative stress and apoptosis. Our previous work showed that, in the absence of cytosolic GPx in insulinoma RINm5F cells, an intrinsic activity of 12 lipoxygenase (12(S)-LOX) converts 12S-hydroperoxyeicosatetraenoic acid (12(S)-HpETE) to the bioactive epoxide hepoxilin A(3) (HXA(3)). The aim of the present study was to investigate the effect of HXA(3) on apoptosis as compared to its precursor 12(S)-HpETE and shed light upon the underlying pathways. In contrast to 12(S)-HpETE, which induced apoptosis via the extrinsic pathway, we found HXA(3) not only to prevent it but also to promote cell proliferation. In particular, HXA(3) suppressed the pro-apoptotic BAX and upregulated the anti-apoptotic Bcl-2. Moreover, HXA(3) induced the anti-apoptotic 12(S)-LOX by recruiting heat shock protein 90 (HSP90), another anti-apoptotic protein. Finally, a co-chaperone protein of HSP90, protein phosphatase 5 (PP5), was upregulated by HXA(3), which counteracted oxidative stress-induced apoptosis by dephosphorylating and thus inactivating apoptosis signal-regulating kinase 1 (ASK1). Taken together, these findings suggest that HXA(3) protects insulinoma cells from oxidative stress and, via multiple signaling pathways, prevents them from undergoing apoptosis.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Apoptosis/drug effects , Insulin-Secreting Cells/drug effects , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Immunoblotting , Insulin-Secreting Cells/metabolism , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Cardiovasc Res ; 84(3): 416-24, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19620128

ABSTRACT

AIMS: We have previously shown that cardiac-specific inhibition of NF-kappaB attenuates angiotensin II (AngII)-induced left ventricular (LV) hypertrophy in vivo. We now tested whether NF-kappaB inhibition is able to block LV remodelling upon chronic pressure overload and chronic AngII stimulation. METHODS AND RESULTS: Cardiac-restricted NF-kappaB inhibition was achieved by expression of a stabilized IkappaBalpha mutant (IkappaBalphaDeltaN) in cells with an active alpha-myosin heavy chain (alphaMHC) promoter employing the Cre/lox technique. Upon low-gradient trans-aortic constriction (TAC, gradient 21 +/- 3 mmHg), hypertrophy was induced in both male and female control mice after 4 weeks. At this time, LV hypertrophy was blocked in transgenic (TG) male but not female mice with NF-kappaB inhibition. Amelioration of LV hypertrophy was associated with activation of NF-kappaB by dihydrotestosterone in isolated neonatal cardiomyocytes. LV remodelling was not attenuated by NF-kappaB inhibition after 8 weeks TAC, demonstrated by decreased fractional shortening (FS) in both control and TG mice irrespective of gender. Similar results were obtained when TAC was performed with higher gradients (48 +/- 4 mmHg). In TG mice, FS dropped to similar low levels over the same time course [FS sham, 29 +/- 1% (mean +/- SEM); FS control + 14 days TAC, 13 +/- 3%; FS TG + 14 days TAC, 9 +/- 5%]. Similarly, LV remodelling was accelerated by NF-kappaB inhibition in an AngII-dependent genetic heart failure model (AT1-R(alphaMHC)) associated with significantly increased cardiac fibrosis in double AT1-R(alphaMHC)/TG mice. CONCLUSION: NF-kappaB inhibition attenuates cardiac hypertrophy in a gender-specific manner but does not alter the course of stress-induced LV remodelling, indicating NF-kappaB to be required for adaptive cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/physiopathology , NF-kappa B/metabolism , Sex Characteristics , Ventricular Remodeling/physiology , Angiotensin II/metabolism , Animals , Apoptosis/physiology , Cardiomegaly/pathology , Disease Models, Animal , Female , Fibrosis , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Receptor, Angiotensin, Type 1/physiology , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 105(50): 19762-7, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19073933

ABSTRACT

We analyzed the effect of conditional, alphaMHC-dependent genetic beta-catenin depletion and stabilization on cardiac remodeling following experimental infarct. beta-Catenin depletion significantly improved 4-week survival and left ventricular (LV) function (fractional shortening: CT(Deltaex3-6): 24 +/- 1.9%; beta-cat(Deltaex3-6): 30.2 +/- 1.6%, P < 0.001). beta-Catenin stabilization had opposite effects. No significant changes in adult cardiomyocyte survival or hypertrophy were observed in either transgenic line. Associated with the functional improvement, LV scar cellularity was altered: beta-catenin-depleted mice showed a marked subendocardial and subepicardial layer of small cTnT(pos) cardiomyocytes associated with increased expression of cardiac lineage markers Tbx5 and GATA4. Using a Cre-dependent lacZ reporter gene, we identified a noncardiomyocyte cell population affected by alphaMHC-driven gene recombination localized to these tissue compartments at baseline. These cells were found to be cardiac progenitor cells since they coexpressed markers of proliferation (Ki67) and the cardiomyocyte lineage (alphaMHC, GATA4, Tbx5) but not cardiac Troponin T (cTnT). The cell population overlaps in part with both the previously described c-kit(pos) and stem cell antigen-1 (Sca-1)(pos) precursor cell population but not with the Islet-1(pos) precursor cell pool. An in vitro coculture assay of highly enriched (>95%) Sca-1(pos) cardiac precursor cells from beta-catenin-depleted mice compared to cells isolated from control littermate demonstrated increased differentiation toward alpha-actin(pos) and cTnT(pos) cardiomyocytes after 10 days (CT(Deltaex3-6): 38.0 +/- 1.0% alpha-actin(pos); beta-cat(Deltaex3-6): 49.9 +/- 2.4% alpha-actin(pos), P < 0.001). We conclude that beta-catenin depletion attenuates postinfarct LV remodeling in part through increased differentiation of GATA4(pos)/Sca-1(pos) resident cardiac progenitor cells.


Subject(s)
Myoblasts, Cardiac/physiology , Myocardial Infarction/metabolism , Regeneration , Ventricular Remodeling , beta Catenin/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation , Down-Regulation , Genes, Reporter , Mice , Mice, Transgenic , Myoblasts, Cardiac/pathology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Regeneration/genetics , Ventricular Remodeling/genetics , beta Catenin/genetics , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...