Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37873297

ABSTRACT

During heart development, a well-characterized network of transcription factors initiates cardiac gene expression and defines the precise timing and location of cardiac progenitor specification. However, our understanding of the post-initiation transcriptional events that regulate cardiac gene expression is still incomplete. The PAF1C component Rtf1 is a transcription regulatory protein that modulates pausing and elongation of RNA Pol II, as well as cotranscriptional histone modifications. Here we report that Rtf1 is essential for cardiogenesis in fish and mammals, and that in the absence of Rtf1 activity, cardiac progenitors arrest in an immature state. We found that Rtf1's Plus3 domain, which confers interaction with the transcriptional pausing and elongation regulator Spt5, was necessary for cardiac progenitor formation. ChIP-seq analysis further revealed changes in the occupancy of RNA Pol II around the transcription start site (TSS) of cardiac genes in rtf1 morphants reflecting a reduction in transcriptional pausing. Intriguingly, inhibition of pause release in rtf1 morphants and mutants restored the formation of cardiac cells and improved Pol II occupancy at the TSS of key cardiac genes. Our findings highlight the crucial role that transcriptional pausing plays in promoting normal gene expression levels in a cardiac developmental context.

2.
J Cardiovasc Dev Dis ; 10(5)2023 May 20.
Article in English | MEDLINE | ID: mdl-37233188

ABSTRACT

The PAF1 complex component Rtf1 is an RNA Polymerase II-interacting transcription regulatory protein that promotes transcription elongation and the co-transcriptional monoubiquitination of histone 2B. Rtf1 plays an essential role in the specification of cardiac progenitors from the lateral plate mesoderm during early embryogenesis, but its requirement in mature cardiac cells is unknown. Here, we investigate the importance of Rtf1 in neonatal and adult cardiomyocytes using knockdown and knockout approaches. We demonstrate that loss of Rtf1 activity in neonatal cardiomyocytes disrupts cell morphology and results in a breakdown of sarcomeres. Similarly, Rtf1 ablation in mature cardiomyocytes of the adult mouse heart leads to myofibril disorganization, disrupted cell-cell junctions, fibrosis, and systolic dysfunction. Rtf1 knockout hearts eventually fail and exhibit structural and gene expression defects resembling dilated cardiomyopathy. Intriguingly, we observed that loss of Rtf1 activity causes a rapid change in the expression of key cardiac structural and functional genes in both neonatal and adult cardiomyocytes, suggesting that Rtf1 is continuously required to support expression of the cardiac gene program.

3.
Front Physiol ; 12: 724828, 2021.
Article in English | MEDLINE | ID: mdl-34483974

ABSTRACT

Mitochondria critically regulate a range of cellular processes including bioenergetics, cellular metabolism, apoptosis, and cellular Ca2+ signaling. The voltage-dependent anion channel (VDAC) functions as a passageway for the exchange of ions, including Ca2+, across the outer mitochondrial membrane. In cardiomyocytes, genetic or pharmacological activation of isoform 2 of VDAC (VDAC2) effectively potentiates mitochondrial Ca2+ uptake and suppresses Ca2+ overload-induced arrhythmogenic events. However, molecular mechanisms by which VDAC2 controls mitochondrial Ca2+ transport and thereby influences cardiac rhythmicity remain elusive. Vertebrates express three highly homologous VDAC isoforms. Here, we used the zebrafish tremblor/ncx1h mutant to dissect the isoform-specific roles of VDAC proteins in Ca2+ handling. We found that overexpression of VDAC1 or VDAC2, but not VDAC3, suppresses the fibrillation-like phenotype in zebrafish tremblor/ncx1h mutants. A chimeric approach showed that moieties in the N-terminal half of VDAC are responsible for their divergent functions in cardiac biology. Phylogenetic analysis further revealed that a glutamate at position 73, which was previously described to be an important regulator of VDAC function, is sevolutionarily conserved in VDAC1 and VDAC2, whereas a glutamine occupies position 73 (Q73) of VDAC3. To investigate whether E73/Q73 determines VDAC isoform-specific anti-arrhythmic effect, we mutated E73 to Q in VDAC2 (VDAC2E73Q) and Q73 to E in VDAC3 (VDAC3Q73E). Interestingly, VDAC2E73Q failed to restore rhythmic cardiac contractions in ncx1 deficient hearts, while the Q73E conversion induced a gain of function in VDAC3. In HL-1 cardiomyocytes, VDAC2 knockdown diminished the transfer of Ca2+ from the SR into mitochondria and overexpression of VDAC2 or VDAC3Q73E restored SR-mitochondrial Ca2+ transfer in VDAC2 deficient HL-1 cells, whereas this rescue effect was absent for VDAC3 and drastically compromised for VDAC2E73Q. Collectively, our findings demonstrate a critical role for the evolutionary conserved E73 in determining the anti-arrhythmic effect of VDAC isoforms through modulating Ca2+ cross-talk between the SR and mitochondria in cardiomyocytes.

4.
Cancer Res ; 81(3): 619-633, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33218969

ABSTRACT

Metastases largely rely on hematogenous dissemination of tumor cells via the vascular system and significantly limit prognosis of patients with solid tumors. To colonize distant sites, circulating tumor cells must destabilize the endothelial barrier and transmigrate across the vessel wall. Here we performed a high-content screen to identify drugs that block tumor cell extravasation by testing 3,520 compounds on a transendothelial invasion coculture assay. Hits were further characterized and validated using a series of in vitro assays, a zebrafish model enabling three-dimensional (3D) visualization of tumor cell extravasation, and mouse models of lung metastasis. The initial screen advanced 38 compounds as potential hits, of which, four compounds enhanced endothelial barrier stability while concurrently suppressing tumor cell motility. Two compounds niclosamide and forskolin significantly reduced tumor cell extravasation in zebrafish, and niclosamide drastically impaired metastasis in mice. Because niclosamide had not previously been linked with effects on barrier function, single-cell RNA sequencing uncovered mechanistic effects of the drug on both tumor and endothelial cells. Importantly, niclosamide affected homotypic and heterotypic signaling critical to intercellular junctions, cell-matrix interactions, and cytoskeletal regulation. Proteomic analysis indicated that niclosamide-treated mice also showed reduced levels of kininogen, the precursor to the permeability mediator bradykinin. Our findings designate niclosamide as an effective drug that restricts tumor cell extravasation through modulation of signaling pathways, chemokines, and tumor-endothelial cell interactions. SIGNIFICANCE: A high-content screen identified niclosamide as an effective drug that restricts tumor cell extravasation by enhancing endothelial barrier stability through modulation of molecular signaling, chemokines, and tumor-endothelial cell interactions. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/3/619/F1.large.jpg.


Subject(s)
Colforsin/pharmacology , Endothelium, Vascular , Lung Neoplasms/pathology , Neoplastic Cells, Circulating , Niclosamide/pharmacology , Transendothelial and Transepithelial Migration/drug effects , Animals , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor/methods , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Kininogens/analysis , Male , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Invasiveness , Proteomics , Zebrafish
5.
Front Physiol ; 11: 617492, 2020.
Article in English | MEDLINE | ID: mdl-33424641

ABSTRACT

Mitochondrial Ca2 + uptake influences energy production, cell survival, and Ca2 + signaling. The mitochondrial calcium uniporter, MCU, is the primary route for uptake of Ca2 + into the mitochondrial matrix. We have generated a zebrafish MCU mutant that survives to adulthood and exhibits dramatic cardiac phenotypes resembling cardiomyopathy and sinus arrest. MCU hearts contract weakly and have a smaller ventricle with a thin compact layer and reduced trabecular density. Damaged myofibrils and swollen mitochondria were present in the ventricles of MCU mutants, along with gene expression changes indicative of cell stress and altered cardiac structure and function. Using electrocardiography, we found that MCU hearts display conduction system defects and abnormal rhythm, with extended pauses resembling episodes of sinus arrest. Together, our findings suggest that proper mitochondrial Ca2 + homeostasis is crucial for maintaining a healthy adult heart, and establish the MCU mutant as a useful model for understanding the role of mitochondrial Ca2 + handling in adult cardiac biology.

6.
Org Lett ; 20(19): 6089-6093, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30246538

ABSTRACT

P-Chiral [2.2.1] bicyclic phosphines (HypPhos catalysts) have been applied to reactions between α-alkylallenoates and imines, producing guvacine derivatives. These HypPhos catalysts were assembled from trans-4-hydroxyproline, with the modular nature of the synthesis allowing variations of the exocyclic P and N substituents. Among them, exo-( p-anisyl)-HypPhos was most efficacious for [4 + 2] annulations between ethyl α-methylallenoate and imines. Through this method, ( R)-aplexone was identified as being responsible for the decrease in the cellular levels of cholesterol.


Subject(s)
Imines/chemistry , Naphthalenes/chemistry , Nicotinic Acids/chemistry , Nicotinic Acids/chemical synthesis , Catalysis , Chemistry Techniques, Synthetic , Stereoisomerism , Substrate Specificity
7.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135307

ABSTRACT

The ability to image tissue morphogenesis in real-time and in 3-dimensions (3-D) remains an optical challenge. The advent of light-sheet fluorescence microscopy (LSFM) has advanced developmental biology and tissue regeneration research. In this review, we introduce a LSFM system in which the illumination lens reshapes a thin light-sheet to rapidly scan across a sample of interest while the detection lens orthogonally collects the imaging data. This multiscale strategy provides deep-tissue penetration, high-spatiotemporal resolution, and minimal photobleaching and phototoxicity, allowing in vivo visualization of a variety of tissues and processes, ranging from developing hearts in live zebrafish embryos to ex vivo interrogation of the microarchitecture of optically cleared neonatal hearts. Here, we highlight multiple applications of LSFM and discuss several studies that have allowed better characterization of developmental and pathological processes in multiple models and tissues. These findings demonstrate the capacity of multiscale light-sheet imaging to uncover cardiovascular developmental and regenerative phenomena.


Subject(s)
Heart/diagnostic imaging , Imaging, Three-Dimensional/methods , Intravital Microscopy/methods , Respiratory System/diagnostic imaging , Animals , Animals, Newborn , Embryo, Nonmammalian , Heart/embryology , Heart/growth & development , Imaging, Three-Dimensional/instrumentation , Intravital Microscopy/instrumentation , Light , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Models, Animal , Morphogenesis , Respiratory System/embryology , Respiratory System/growth & development , Time-Lapse Imaging/instrumentation , Time-Lapse Imaging/methods
8.
Cell Rep ; 22(5): 1211-1224, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29386109

ABSTRACT

Given its role as the source of definitive hematopoietic cells, we sought to determine whether mutations initiated in the hemogenic endothelium would yield hematopoietic abnormalities or malignancies. Here, we find that endothelium-specific transposon mutagenesis in mice promotes hematopoietic pathologies that are both myeloid and lymphoid in nature. Frequently mutated genes included previously recognized cancer drivers and additional candidates, such as Pi4ka, a lipid kinase whose mutation was found to promote myeloid and erythroid dysfunction. Subsequent validation experiments showed that targeted inactivation of the Pi4ka catalytic domain or reduction in mRNA expression inhibited myeloid and erythroid cell differentiation in vitro and promoted anemia in vivo through a mechanism involving deregulation of AKT, MAPK, SRC, and JAK-STAT signaling. Finally, we provide evidence linking PI4KAP2, previously considered a pseudogene, to human myeloid and erythroid leukemia.


Subject(s)
Erythropoiesis/physiology , Leukemia/genetics , Minor Histocompatibility Antigens/genetics , Myelopoiesis/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Cell Differentiation/genetics , Hemangioblasts/cytology , Hemangioblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/metabolism , Mutagenesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Zebrafish
9.
Elife ; 62017 08 19.
Article in English | MEDLINE | ID: mdl-28826496

ABSTRACT

Altered Ca2+ handling is often present in diseased hearts undergoing structural remodeling and functional deterioration. However, whether Ca2+ directly regulates sarcomere structure has remained elusive. Using a zebrafish ncx1 mutant, we explored the impacts of impaired Ca2+ homeostasis on myofibril integrity. We found that the E3 ubiquitin ligase murf1 is upregulated in ncx1-deficient hearts. Intriguingly, knocking down murf1 activity or inhibiting proteasome activity preserved myofibril integrity, revealing a MuRF1-mediated proteasome degradation mechanism that is activated in response to abnormal Ca2+ homeostasis. Furthermore, we detected an accumulation of the murf1 regulator FoxO in the nuclei of ncx1-deficient cardiomyocytes. Overexpression of FoxO in wild type cardiomyocytes induced murf1 expression and caused myofibril disarray, whereas inhibiting Calcineurin activity attenuated FoxO-mediated murf1 expression and protected sarcomeres from degradation in ncx1-deficient hearts. Together, our findings reveal a novel mechanism by which Ca2+ overload disrupts myofibril integrity by activating a Calcineurin-FoxO-MuRF1-proteosome signaling pathway.


Subject(s)
Calcineurin/genetics , Calcium/metabolism , Forkhead Box Protein O1/genetics , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Ubiquitin-Protein Ligases/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Calcineurin/metabolism , Calcium Signaling , Embryo, Nonmammalian , Forkhead Box Protein O1/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/ultrastructure , Myocytes, Cardiac/ultrastructure , Myofibrils/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Zebrafish , Zebrafish Proteins/metabolism
10.
Dev Biol ; 421(2): 139-148, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27940156

ABSTRACT

Tbx20 is a T-box transcription factor that plays essential roles in the development and maintenance of the heart. Although it is expressed by cardiac progenitors in all species examined, an involvement of Tbx20 in cardiac progenitor formation in vertebrates has not been previously described. Here we report the identification of a zebrafish tbx20 mutation that results in an inactive, truncated protein lacking any functional domains. The cardiac progenitor population is strongly diminished in this mutant, leading to the formation of a small, stretched-out heart. We found that overexpression of Tbx20 results in an enlarged heart with significantly more cardiomyocytes. Interestingly, this increase in cell number is caused by both enhanced cardiac progenitor cell formation and the proliferation of differentiated cardiomyocytes, and is dependent upon the activity of Tbx20's T-box and transcription activation domains. Together, our findings highlight a previously unappreciated role for Tbx20 in promoting cardiac progenitor formation in vertebrates and reveal a novel function for its activation domain in cardiac cell proliferation during embryogenesis.


Subject(s)
Myocytes, Cardiac/cytology , Organogenesis , Stem Cells/cytology , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Apoptosis/genetics , Base Sequence , Cell Count , Cell Proliferation , Cloning, Molecular , Codon, Nonsense/genetics , DNA/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Mutation/genetics , Myocardium/cytology , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Protein Binding/genetics , Protein Domains , Stem Cells/metabolism , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Transcriptional Activation/genetics , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
11.
J Biol Chem ; 291(52): 26636-26646, 2016 Dec 23.
Article in English | MEDLINE | ID: mdl-27815504

ABSTRACT

Calmodulin (CaM) is a Ca2+ binding protein modulating multiple targets, several of which are associated with cardiac pathophysiology. Recently, CaM mutations were linked to heart arrhythmia. CaM is crucial for cell growth and viability, yet the effect of the arrhythmogenic CaM mutations on cell viability, as well as heart rhythm, remains unknown, and only a few targets with relevance for heart physiology have been analyzed for their response to mutant CaM. We show that the arrhythmia-associated CaM mutants support growth and viability of DT40 cells in the absence of WT CaM except for the long QT syndrome mutant CaM D129G. Of the six CaM mutants tested (N53I, F89L, D95V, N97S, D129G, and F141L), three showed a decreased activation of Ca2+/CaM-dependent kinase II, most prominently the D129G CaM mutation, which was incapable of stimulating Thr286 autophosphorylation. Furthermore, the CaM D129G mutation led to bradycardia in zebrafish and an arrhythmic phenotype in a subset of the analyzed zebrafish.


Subject(s)
Arrhythmias, Cardiac/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/genetics , Cell Proliferation/genetics , Mutation/genetics , Tachycardia, Ventricular/pathology , Animals , Calcium/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , Cells, Cultured , Humans , Long QT Syndrome/etiology , Long QT Syndrome/metabolism , Long QT Syndrome/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Protein Conformation , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
12.
J Cardiovasc Dev Dis ; 3(2)2016 Apr 09.
Article in English | MEDLINE | ID: mdl-27148546

ABSTRACT

Cardiac transcription factors orchestrate the complex cellular and molecular events required to produce a functioning heart. Misregulation of the cardiac transcription program leads to embryonic developmental defects and is associated with human congenital heart diseases. Recent studies have expanded our understanding of the regulation of cardiac gene expression at an additional layer, involving the coordination of epigenetic and transcriptional regulators. In this review, we highlight and discuss discoveries made possible by the genetic and embryological tools available in the zebrafish model organism, with a focus on the novel functions of cardiac transcription factors and epigenetic and transcriptional regulatory proteins during cardiogenesis.

13.
J Clin Invest ; 126(1): 195-206, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26619120

ABSTRACT

RNA splicing is a major contributor to total transcriptome complexity; however, the functional role and regulation of splicing in heart failure remain poorly understood. Here, we used a total transcriptome profiling and bioinformatic analysis approach and identified a muscle-specific isoform of an RNA splicing regulator, RBFox1 (also known as A2BP1), as a prominent regulator of alternative RNA splicing during heart failure. Evaluation of developing murine and zebrafish hearts revealed that RBFox1 is induced during postnatal cardiac maturation. However, we found that RBFox1 is markedly diminished in failing human and mouse hearts. In a mouse model, RBFox1 deficiency in the heart promoted pressure overload-induced heart failure. We determined that RBFox1 is a potent regulator of RNA splicing and is required for a conserved splicing process of transcription factor MEF2 family members that yields different MEF2 isoforms with differential effects on cardiac hypertrophic gene expression. Finally, induction of RBFox1 expression in murine pressure overload models substantially attenuated cardiac hypertrophy and pathological manifestations. Together, this study identifies regulation of RNA splicing by RBFox1 as an important player in transcriptome reprogramming during heart failure that influence pathogenesis of the disease.


Subject(s)
Cardiomegaly/etiology , Heart Failure/etiology , RNA Splicing , RNA-Binding Proteins/physiology , Animals , Cardiomegaly/genetics , Heart Failure/genetics , Humans , MEF2 Transcription Factors/genetics , Mice , RNA Splicing Factors , Transcriptome
14.
Dev Biol ; 404(2): 103-12, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26086691

ABSTRACT

Cardiac neural crest cells are essential for outflow tract remodeling in animals with divided systemic and pulmonary circulatory systems, but their contributions to cardiac development in animals with a single-loop circulatory system are less clear. Here we genetically labeled neural crest cells and examined their contribution to the developing zebrafish heart. We identified two populations of neural crest cells that contribute to distinct compartments of zebrafish cardiovascular system at different developmental stages. A stream of neural crest cells migrating through pharyngeal arches 1 and 2 integrates into the myocardium of the primitive heart tube between 24 and 30 h post fertilization and gives rise to cardiomyocytes. A second wave of neural crest cells migrating along aortic arch 6 envelops the endothelium of the ventral aorta and invades the bulbus arteriosus after three days of development. Interestingly, while inhibition of FGF signaling has no effect on the integration of neural crest cells to the primitive heart tube, it prevents these cells from contributing to the outflow tract, demonstrating disparate responses of neural crest cells to FGF signaling. Furthermore, neural crest ablation in zebrafish leads to multiple cardiac defects, including reduced heart rate, defective myocardial maturation and a failure to recruit progenitor cells from the second heart field. These findings add to our understanding of the contribution of neural crest cells to the developing heart and provide insights into the requirement for these cells in cardiac maturation.


Subject(s)
Fibroblast Growth Factors/antagonists & inhibitors , Heart Defects, Congenital/embryology , Heart/embryology , Neural Crest/cytology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Aorta/cytology , Cell Differentiation , Cell Movement , Coronary Vessels/cytology , Coronary Vessels/embryology , Myocytes, Cardiac/cytology
15.
Elife ; 42015 Jan 15.
Article in English | MEDLINE | ID: mdl-25588501

ABSTRACT

Tightly regulated Ca(2+) homeostasis is a prerequisite for proper cardiac function. To dissect the regulatory network of cardiac Ca(2+) handling, we performed a chemical suppressor screen on zebrafish tremblor embryos, which suffer from Ca(2+) extrusion defects. Efsevin was identified based on its potent activity to restore coordinated contractions in tremblor. We show that efsevin binds to VDAC2, potentiates mitochondrial Ca(2+) uptake and accelerates the transfer of Ca(2+) from intracellular stores into mitochondria. In cardiomyocytes, efsevin restricts the temporal and spatial boundaries of Ca(2+) sparks and thereby inhibits Ca(2+) overload-induced erratic Ca(2+) waves and irregular contractions. We further show that overexpression of VDAC2 recapitulates the suppressive effect of efsevin on tremblor embryos whereas VDAC2 deficiency attenuates efsevin's rescue effect and that VDAC2 functions synergistically with MCU to suppress cardiac fibrillation in tremblor. Together, these findings demonstrate a critical modulatory role for VDAC2-dependent mitochondrial Ca(2+) uptake in the regulation of cardiac rhythmicity.


Subject(s)
Calcium/metabolism , Heart Rate , Heart/physiopathology , Mitochondria/metabolism , Voltage-Dependent Anion Channel 2/metabolism , Zebrafish Proteins/metabolism , Zebrafish/physiology , Amino Acid Sequence , Animals , Calcium Signaling/drug effects , Embryo, Mammalian/metabolism , Heart Rate/drug effects , Mitochondria/drug effects , Molecular Sequence Data , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Video Recording , Voltage-Dependent Anion Channel 2/chemistry , Zebrafish/embryology , Zebrafish Proteins/chemistry
16.
Genes Dev ; 28(22): 2547-63, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25403183

ABSTRACT

Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the nucleus, highlighting critical functions of Sufu in the nucleus. Through a proteomic approach, we identified several Sufu-interacting proteins, including p66ß (a member of the NuRD [nucleosome remodeling and histone deacetylase] repressor complex) and Mycbp (a Myc-binding protein). p66ß negatively and Mycbp positively regulate Hh signaling in cell-based assays and zebrafish. They function downstream from the membrane receptors, Patched and Smoothened, and the primary cilium. Sufu, p66ß, Mycbp, and Gli are also detected on the promoters of Hh targets in a dynamic manner. Our results support a new model of Hh signaling in the nucleus. Sufu recruits p66ß to block Gli-mediated Hh target gene expression. Meanwhile, Mycbp forms a complex with Gli and Sufu without Hh stimulation but remains inactive. Hh pathway activation leads to dissociation of Sufu/p66ß from Gli, enabling Mycbp to promote Gli protein activity and Hh target gene expression. These studies provide novel insight into how Sufu controls Hh signaling in the nucleus.


Subject(s)
Gene Expression Regulation , Hedgehog Proteins/physiology , Repressor Proteins/metabolism , Salivary alpha-Amylases/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Knockdown Techniques , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mutation , NIH 3T3 Cells , Protein Binding , Proteomics , Repressor Proteins/genetics , Salivary alpha-Amylases/genetics , Zebrafish/genetics , Zinc Finger Protein GLI1
17.
J Biol Chem ; 289(33): 23200-23208, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24962575

ABSTRACT

Oxidative stress has been implicated in cardiac arrhythmia, although a causal relationship remains undefined. We have recently demonstrated a marked up-regulation of NADPH oxidase isoform 4 (NOX4) in patients with atrial fibrillation, which is accompanied by overproduction of reactive oxygen species (ROS). In this study, we investigated the impact on the cardiac phenotype of NOX4 overexpression in zebrafish. One-cell stage embryos were injected with NOX4 RNA prior to video recording of a GFP-labeled (myl7:GFP zebrafish line) beating heart in real time at 24-31 h post-fertilization. Intriguingly, NOX4 embryos developed cardiac arrhythmia that is characterized by irregular heartbeats. When quantitatively analyzed by an established LQ-1 program, the NOX4 embryos displayed much more variable beat-to-beat intervals (mean S.D. of beat-to-beat intervals was 0.027 s/beat in control embryos versus 0.038 s/beat in NOX4 embryos). Both the phenotype and the increased ROS in NOX4 embryos were attenuated by NOX4 morpholino co-injection, treatments of the embryos with polyethylene glycol-conjugated superoxide dismutase, or NOX4 inhibitors fulvene-5, 6-dimethylamino-fulvene, and proton sponge blue. Injection of NOX4-P437H mutant RNA had no effect on the cardiac phenotype or ROS production. In addition, phosphorylation of calcium/calmodulin-dependent protein kinase II was increased in NOX4 embryos but diminished by polyethylene glycol-conjugated superoxide dismutase, whereas its inhibitor KN93 or AIP abolished the arrhythmic phenotype. Taken together, our data for the first time uncover a novel pathway that underlies the development of cardiac arrhythmia, namely NOX4 activation, subsequent NOX4-specific NADPH-driven ROS production, and redox-sensitive CaMKII activation. These findings may ultimately lead to novel therapeutics targeting cardiac arrhythmia.


Subject(s)
Arrhythmias, Cardiac/enzymology , Myocardial Contraction , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Arrhythmias, Cardiac/embryology , Arrhythmias, Cardiac/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , NADPH Oxidases/genetics , Phenotype , Zebrafish/genetics , Zebrafish Proteins/genetics
18.
J Biol Chem ; 289(18): 12566-77, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24627492

ABSTRACT

In recent years, there has been a vast increase in structural and functional understanding of VDAC1, but VDAC2 and -3 have been understudied despite having many unique phenotypes. One reason for the paucity of structural and biochemical characterization of the VDAC2 and -3 isoforms stems from the inability of obtaining purified, functional protein. Here we demonstrate the expression, isolation, and basic characterization of zebrafish VDAC2 (zfVDAC2). Further, we resolved the structure of zfVDAC2 at 2.8 Šresolution, revealing a crystallographic dimer. The dimer orientation was confirmed in solution by double electron-electron resonance spectroscopy and by cross-linking experiments disclosing a dimer population of ∼20% in lauryldimethine amine oxide detergent micelles, whereas in lipidic bicelles a higher population of dimeric and higher order oligomers species were observed. The present study allows for a more accurate structural comparison between VDAC2 and its better-studied counterpart VDAC1.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Protein Multimerization , Voltage-Dependent Anion Channel 2/chemistry , Zebrafish Proteins/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Electric Conductivity , Electrophoresis, Polyacrylamide Gel , Lipid Bilayers/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Secondary , Sequence Homology, Amino Acid , Static Electricity , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
Am J Physiol Heart Circ Physiol ; 305(11): H1624-38, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24077883

ABSTRACT

Myocyte hypertrophy antecedent to heart failure involves changes in global gene expression, although the preceding mechanisms to coordinate DNA accessibility on a genomic scale are unknown. Chromatin-associated proteins alter chromatin structure by changing their association with DNA, thereby altering the gene expression profile. Little is known about the global changes in chromatin subproteomes that accompany heart failure, and the mechanisms by which these proteins alter chromatin structure. The present study tests the fundamental hypothesis that cardiac growth and plasticity in the setting of disease recapitulates conserved developmental chromatin remodeling events. We used quantitative proteomics to identify chromatin-associated proteins extracted via detergent and to quantify changes in their abundance during disease. Our study identified 321 proteins in this subproteome, demonstrating it to have modest conservation (37%) with that revealed using strong acid. Of these proteins, 176 exhibited altered expression during cardiac hypertrophy and failure; we conducted extensive functional characterization of one of these proteins, Nucleolin. Morpholino-based knockdown of nucleolin nearly abolished protein expression but surprisingly had little impact on gross morphological development. However, hearts of fish lacking Nucleolin displayed severe developmental impairment, abnormal chamber patterning and functional deficits, ostensibly due to defects in cardiac looping and myocyte differentiation. The mechanisms underlying these defects involve perturbed bone morphogenetic protein 4 expression, decreased rRNA transcription, and a shift to more heterochromatic chromatin. This study reports the quantitative analysis of a new chromatin subproteome in the normal and diseased mouse heart. Validation studies in the complementary model system of zebrafish examine the role of Nucleolin to orchestrate genomic reprogramming events shared between development and disease.


Subject(s)
Cardiomegaly/metabolism , Chromatin/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Proteomics , RNA-Binding Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Chromatin Assembly and Disassembly , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Gene Knockdown Techniques , Heart Failure/genetics , Heart Failure/pathology , Male , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/pathology , Phosphoproteins/genetics , Proteomics/methods , RNA, Ribosomal/metabolism , RNA-Binding Proteins/genetics , Rats , Signal Transduction , Time Factors , Transcription, Genetic , Zebrafish , Zebrafish Proteins/genetics , Nucleolin
20.
Cell ; 150(3): 590-605, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863011

ABSTRACT

Endothelium in embryonic hematopoietic tissues generates hematopoietic stem/progenitor cells; however, it is unknown how its unique potential is specified. We show that transcription factor Scl/Tal1 is essential for both establishing the hematopoietic transcriptional program in hemogenic endothelium and preventing its misspecification to a cardiomyogenic fate. Scl(-/-) embryos activated a cardiac transcriptional program in yolk sac endothelium, leading to the emergence of CD31+Pdgfrα+ cardiogenic precursors that generated spontaneously beating cardiomyocytes. Ectopic cardiogenesis was also observed in Scl(-/-) hearts, where the disorganized endocardium precociously differentiated into cardiomyocytes. Induction of mosaic deletion of Scl in Scl(fl/fl)Rosa26Cre-ER(T2) embryos revealed a cell-intrinsic, temporal requirement for Scl to prevent cardiomyogenesis from endothelium. Scl(-/-) endothelium also upregulated the expression of Wnt antagonists, which promoted rapid cardiomyocyte differentiation of ectopic cardiogenic cells. These results reveal unexpected plasticity in embryonic endothelium such that loss of a single master regulator can induce ectopic cardiomyogenesis from endothelial cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelium, Vascular/embryology , Heart/embryology , Proto-Oncogene Proteins/metabolism , Animals , Cadherins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Gene Expression Regulation, Developmental , Hemangioblasts , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , LIM-Homeodomain Proteins/metabolism , Mesoderm/metabolism , Mice , Myocytes, Cardiac/cytology , Placenta/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Receptor, Platelet-Derived Growth Factor alpha/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/metabolism , Yolk Sac/blood supply
SELECTION OF CITATIONS
SEARCH DETAIL
...