57 results on '"Small EM"'
Search Results
2. Multi-omics analysis reveals the dynamic interplay between Vero host chromatin structure and function during vaccinia virus infection.
- Author
-
Venu V, Roth C, Adikari SH, Small EM, Starkenburg SR, Sanbonmatsu KY, and Steadman CR
- Subjects
- Animals, Chlorocebus aethiops, Vero Cells, Vaccinia virology, Vaccinia immunology, Host-Pathogen Interactions genetics, Multiomics, Chromatin metabolism, Chromatin genetics, Vaccinia virus genetics, Vaccinia virus physiology
- Abstract
The genome folds into complex configurations and structures thought to profoundly impact its function. The intricacies of this dynamic structure-function relationship are not well understood particularly in the context of viral infection. To unravel this interplay, here we provide a comprehensive investigation of simultaneous host chromatin structural (via Hi-C and ATAC-seq) and functional changes (via RNA-seq) in response to vaccinia virus infection. Over time, infection significantly impacts global and local chromatin structure by increasing long-range intra-chromosomal interactions and B compartmentalization and by decreasing chromatin accessibility and inter-chromosomal interactions. Local accessibility changes are independent of broad-scale chromatin compartment exchange (~12% of the genome), underscoring potential independent mechanisms for global and local chromatin reorganization. While infection structurally condenses the host genome, there is nearly equal bidirectional differential gene expression. Despite global weakening of intra-TAD interactions, functional changes including downregulated immunity genes are associated with alterations in local accessibility and loop domain restructuring. Therefore, chromatin accessibility and local structure profiling provide impactful predictions for host responses and may improve development of efficacious anti-viral counter measures including the optimization of vaccine design., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. p53 Regulates the Extent of Fibroblast Proliferation and Fibrosis in Left Ventricle Pressure Overload.
- Author
-
Liu X, Burke RM, Lighthouse JK, Baker CD, Dirkx RA Jr, Kang B, Chakraborty Y, Mickelsen DM, Twardowski JJ, Mello SS, Ashton JM, and Small EM
- Subjects
- Mice, Animals, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Fibrosis, Fibroblasts metabolism, Cell Proliferation, Myocardium metabolism, Heart Ventricles pathology, Cicatrix metabolism
- Abstract
Background: Cardiomyopathy is characterized by the pathological accumulation of resident cardiac fibroblasts that deposit ECM (extracellular matrix) and generate a fibrotic scar. However, the mechanisms that control the timing and extent of cardiac fibroblast proliferation and ECM production are not known, hampering the development of antifibrotic strategies to prevent heart failure., Methods: We used the Tcf21 (transcription factor 21)
MerCreMer mouse line for fibroblast-specific lineage tracing and p53 (tumor protein p53) gene deletion. We characterized cardiac physiology and used single-cell RNA-sequencing and in vitro studies to investigate the p53-dependent mechanisms regulating cardiac fibroblast cell cycle and fibrosis in left ventricular pressure overload induced by transaortic constriction., Results: Cardiac fibroblast proliferation occurs primarily between days 7 and 14 following transaortic constriction in mice, correlating with alterations in p53-dependent gene expression. p53 deletion in fibroblasts led to a striking accumulation of Tcf21-lineage cardiac fibroblasts within the normal proliferative window and precipitated a robust fibrotic response to left ventricular pressure overload. However, excessive interstitial and perivascular fibrosis does not develop until after cardiac fibroblasts exit the cell cycle. Single-cell RNA sequencing revealed p53 null fibroblasts unexpectedly express lower levels of genes encoding important ECM proteins while they exhibit an inappropriately proliferative phenotype. in vitro studies establish a role for p53 in suppressing the proliferative fibroblast phenotype, which facilitates the expression and secretion of ECM proteins. Importantly, Cdkn2a (cyclin-dependent kinase inhibitor 2a) expression and the p16Ink4a -retinoblastoma cell cycle control pathway is induced in p53 null cardiac fibroblasts, which may eventually contribute to cell cycle exit and fulminant scar formation., Conclusions: This study reveals a mechanism regulating cardiac fibroblast accumulation and ECM secretion, orchestrated in part by p53-dependent cell cycle control that governs the timing and extent of fibrosis in left ventricular pressure overload., Competing Interests: Disclosures None.- Published
- 2023
- Full Text
- View/download PDF
4. A screen for histone mutations that affect quiescence in S. cerevisiae.
- Author
-
Small EM and Osley MA
- Subjects
- Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Cell Division, Chromatin genetics, Chromatin metabolism, Mutation, Histones genetics, Histones metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Quiescence or G0 is a reversible state in which cells cease division but retain the ability to resume proliferation. Quiescence occurs in all organisms and is essential for stem cell maintenance and tissue renewal. It is also related to chronological lifespan (CLS)-the survival of postmitotic quiescent cells (Q cells) over time-and thus contributes to longevity. Important questions remain regarding the mechanisms that control entry into quiescence, maintenance of quiescence and re-entry of Q cells into the cell cycle. S. cerevisiae has emerged as an excellent organism in which to address these questions because of the ease in which Q cells can be isolated. Following entry into G0, yeast cells remain viable for an extended period and can re-enter the cell cycle when exposed to growth-promoting signals. Histone acetylation is lost during the formation of Q cells and chromatin becomes highly condensed. This unique chromatin landscape regulates quiescence-specific transcriptional repression and has been linked to the formation and maintenance of Q cells. To ask whether other chromatin features regulate quiescence, we conducted two comprehensive screens of histone H3 and H4 mutants and identified mutants that show either altered quiescence entry or CLS. Examination of several quiescence entry mutants found that none of the mutants retain histone acetylation in Q cells but show differences in chromatin condensation. A comparison of H3 and H4 mutants with altered CLS to those with altered quiescence entry found that chromatin plays both overlapping and independent roles in the continuum of the quiescence program., (© 2023 Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
5. Cardiac pericytes mediate the remodeling response to myocardial infarction.
- Author
-
Quijada P, Park S, Zhao P, Kolluri KS, Wong D, Shih KD, Fang K, Pezhouman A, Wang L, Daraei A, Tran MD, Rathbun EM, Burgos Villar KN, Garcia-Hernandez ML, Pham TT, Lowenstein CJ, Iruela-Arispe ML, Carmichael ST, Small EM, and Ardehali R
- Subjects
- Mice, Animals, Heart, Fibrosis, Extracellular Matrix metabolism, Ventricular Remodeling genetics, Myocardium metabolism, Pericytes metabolism, Myocardial Infarction metabolism
- Abstract
Despite the prevalence of pericytes in the microvasculature of the heart, their role during ischemia-induced remodeling remains unclear. We used multiple lineage-tracing mouse models and found that pericytes migrated to the injury site and expressed profibrotic genes, coinciding with increased vessel leakage after myocardial infarction (MI). Single-cell RNA-Seq of cardiac pericytes at various time points after MI revealed the temporally regulated induction of genes related to vascular permeability, extracellular matrix production, basement membrane degradation, and TGF-β signaling. Deleting TGF-β receptor 1 in chondroitin sulfate proteoglycan 4-expressing (Cspg4-expressing) cells reduced fibrosis following MI, leading to a transient improvement in the cardiac ejection fraction. Furthermore, genetic ablation of Cspg4-expressing cells resulted in excessive vascular permeability, a decline in cardiac function, and increased mortality in the second week after MI. These data reveal an essential role for cardiac pericytes in the control of vascular homeostasis and the fibrotic response after acute ischemic injury, information that will help guide the development of novel strategies to preserve vascular integrity and attenuate pathological cardiac remodeling.
- Published
- 2023
- Full Text
- View/download PDF
6. Cell-Based Phenotypic Screen for Antifibrotic Compounds Targets Eicosanoid Metabolism.
- Author
-
Lighthouse JK and Small EM
- Subjects
- Humans, Fibrosis, Extracellular Matrix metabolism, Myocardium metabolism, Eicosanoids metabolism
- Published
- 2023
- Full Text
- View/download PDF
7. SIRT6 Mitigates Heart Failure With Preserved Ejection Fraction in Diabetes.
- Author
-
Wu X, Liu H, Brooks A, Xu S, Luo J, Steiner R, Mickelsen DM, Moravec CS, Jeffrey AD, Small EM, and Jin ZG
- Subjects
- Humans, Mice, Animals, Stroke Volume physiology, PPAR gamma, Disease Models, Animal, Lipids, Heart Failure metabolism, Diabetes Mellitus, Experimental, Sirtuins genetics
- Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) is a growing health problem without effective therapies. Epidemiological studies indicate that diabetes is a strong risk factor for HFpEF, and about 45% of patients with HFpEF are suffering from diabetes, yet the underlying mechanisms remain elusive., Methods: Using a combination of echocardiography, hemodynamics, RNA-sequencing, molecular biology, in vitro and in vivo approaches, we investigated the roles of SIRT6 (sirtuin 6) in regulation of endothelial fatty acid (FA) transport and HFpEF in diabetes., Results: We first observed that endothelial SIRT6 expression was markedly diminished in cardiac tissues from heart failure patients with diabetes. We then established an experimental mouse model of HFpEF in diabetes induced by a combination of the long-term high-fat diet feeding and a low-dose streptozocin challenge. We also generated a unique humanized SIRT6 transgenic mouse model, in which a single copy of human SIRT6 transgene was engineered at mouse Rosa26 locus and conditionally induced with the Cre-loxP technology. We found that genetically restoring endothelial SIRT6 expression in the diabetic mice ameliorated diastolic dysfunction concurrently with decreased cardiac lipid accumulation. SIRT6 gain- or loss-of-function studies showed that SIRT6 downregulated endothelial FA uptake. Mechanistically, SIRT6 suppressed endothelial expression of PPARγ through SIRT6-dependent deacetylation of histone H3 lysine 9 around PPARγ promoter region; and PPARγ reduction mediated SIRT6-dependent inhibition of endothelial FA uptake. Importantly, oral administration of small molecule SIRT6 activator MDL-800 to diabetic mice mitigated cardiac lipid accumulation and diastolic dysfunction., Conclusions: The impairment of endothelial SIRT6 expression links diabetes to HFpEF through the alteration of FA transport across the endothelial barrier. Genetic and pharmacological strategies that restored endothelial SIRT6 function in mice with diabetes alleviated experimental HFpEF by limiting FA uptake and improving cardiac metabolism, thus warranting further clinical evaluation.
- Published
- 2022
- Full Text
- View/download PDF
8. Loss of Nuclear Envelope Integrity and Increased Oxidant Production Cause DNA Damage in Adult Hearts Deficient in PKP2: A Molecular Substrate of ARVC.
- Author
-
Pérez-Hernández M, van Opbergen CJM, Bagwan N, Vissing CR, Marrón-Liñares GM, Zhang M, Torres Vega E, Sorrentino A, Drici L, Sulek K, Zhai R, Hansen FB, Christensen AH, Boesgaard S, Gustafsson F, Rossing K, Small EM, Davies MJ, Rothenberg E, Sato PY, Cerrone M, Jensen THL, Qvortrup K, Bundgaard H, Delmar M, and Lundby A
- Subjects
- Adult, Animals, DNA Damage, Humans, Hydrogen Peroxide, Mice, Mutation, Myocytes, Cardiac metabolism, Nuclear Envelope metabolism, Nuclear Envelope pathology, Oxidants metabolism, Stroke Volume, Ventricular Function, Left, Arrhythmogenic Right Ventricular Dysplasia pathology, Induced Pluripotent Stem Cells metabolism, Plakophilins genetics, Plakophilins metabolism
- Abstract
Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized by high propensity to life-threatening arrhythmias and progressive loss of heart muscle. More than 40% of reported genetic variants linked to ARVC reside in the PKP2 gene, which encodes the PKP2 protein (plakophilin-2)., Methods: We describe a comprehensive characterization of the ARVC molecular landscape as determined by high-resolution mass spectrometry, RNA sequencing, and transmission electron microscopy of right ventricular biopsy samples obtained from patients with ARVC with PKP2 mutations and left ventricular ejection fraction >45%. Samples from healthy relatives served as controls. The observations led to experimental work using multiple imaging and biochemical techniques in mice with a cardiac-specific deletion of Pkp2 studied at a time of preserved left ventricular ejection fraction and in human induced pluripotent stem cell-derived PKP2-deficient myocytes., Results: Samples from patients with ARVC present a loss of nuclear envelope integrity, molecular signatures indicative of increased DNA damage, and a deficit in transcripts coding for proteins in the electron transport chain. Mice with a cardiac-specific deletion of Pkp2 also present a loss of nuclear envelope integrity, which leads to DNA damage and subsequent excess oxidant production (O
2 .- and H2 O2 ), the latter increased further under mechanical stress (isoproterenol or exercise). Increased oxidant production and DNA damage is recapitulated in human induced pluripotent stem cell-derived PKP2-deficient myocytes. Furthermore, PKP2-deficient cells release H2 O2 into the extracellular environment, causing DNA damage and increased oxidant production in neighboring myocytes in a paracrine manner. Treatment with honokiol increases SIRT3 (mitochondrial nicotinamide adenine dinucleotide-dependent protein deacetylase sirtuin-3) activity, reduces oxidant levels and DNA damage in vitro and in vivo, reduces collagen abundance in the right ventricular free wall, and has a protective effect on right ventricular function., Conclusions: Loss of nuclear envelope integrity and subsequent DNA damage is a key substrate in the molecular pathology of ARVC. We show transcriptional downregulation of proteins of the electron transcript chain as an early event in the molecular pathophysiology of the disease (before loss of left ventricular ejection fraction <45%), which associates with increased oxidant production (O2 .- and H2 O2 ). We propose therapies that limit oxidant formation as a possible intervention to restrict DNA damage in ARVC.- Published
- 2022
- Full Text
- View/download PDF
9. Transcriptional regulation of cardiac fibroblast phenotypic plasticity.
- Author
-
Burgos Villar KN, Liu X, and Small EM
- Abstract
Cardiac fibroblasts play critical roles in the maintenance of cardiac structure and the response to cardiac insult. Extracellular matrix deposition by activated resident cardiac fibroblasts, called myofibroblasts, is an essential wound healing response. However, persistent fibroblast activation contributes to pathological fibrosis and cardiac chamber stiffening, which can cause diastolic dysfunction, heart failure, and initiate lethal arrhythmias. The dynamic and phenotypically plastic nature of cardiac fibroblasts is governed in part by the transcriptional regulation of genes encoding extracellular matrix molecules. Understanding how fibroblasts integrate various biomechanical cues into a precise transcriptional response may uncover therapeutic strategies to prevent fibrosis. Here, we provide an overview of the recent literature on transcriptional control of cardiac fibroblast plasticity and fibrosis, with a focus on canonical and non-canonical TGF-β signaling, biomechanical regulation of Hippo/YAP and Rho/MRTF signaling, and metabolic and epigenetic control of fibroblast activation., Competing Interests: Declaration of Interests: None
- Published
- 2022
- Full Text
- View/download PDF
10. Characterizing Neonatal Heart Maturation, Regeneration, and Scar Resolution Using Spatial Transcriptomics.
- Author
-
Misra A, Baker CD, Pritchett EM, Burgos Villar KN, Ashton JM, and Small EM
- Abstract
The neonatal mammalian heart exhibits a remarkable regenerative potential, which includes fibrotic scar resolution and the generation of new cardiomyocytes. To investigate the mechanisms facilitating heart repair after apical resection in neonatal mice, we conducted bulk and spatial transcriptomic analyses at regenerative and non-regenerative timepoints. Importantly, spatial transcriptomics provided near single-cell resolution, revealing distinct domains of atrial and ventricular myocardium that exhibit dynamic phenotypic alterations during postnatal heart maturation. Spatial transcriptomics also defined the cardiac scar, which transitions from a proliferative to secretory phenotype as the heart loses regenerative potential. The resolving scar is characterized by spatially and temporally restricted programs of inflammation, epicardium expansion and extracellular matrix production, metabolic reprogramming, lipogenic scar extrusion, and cardiomyocyte restoration. Finally, this study revealed the emergence of a regenerative border zone defined by immature cardiomyocyte markers and the robust expression of Sprr1a. Taken together, our study defines the spatially and temporally restricted gene programs that underlie neonatal heart regeneration and provides insight into cardio-restorative mechanisms supporting scar resolution.
- Published
- 2021
- Full Text
- View/download PDF
11. Coordination of endothelial cell positioning and fate specification by the epicardium.
- Author
-
Quijada P, Trembley MA, Misra A, Myers JA, Baker CD, Pérez-Hernández M, Myers JR, Dirkx RA Jr, Cohen ED, Delmar M, Ashton JM, and Small EM
- Subjects
- Animals, Chemokines, Coronary Vessels metabolism, Embryo, Mammalian, Epithelial-Mesenchymal Transition, Gene Expression, Heart, Intercellular Signaling Peptides and Proteins, Mice, Mice, Inbred C57BL, Nerve Tissue Proteins, Nuclear Proteins, Pericardium embryology, Serum Response Factor, Signal Transduction, Trans-Activators, Transcription Factors metabolism, Transcriptome, Endothelial Cells cytology, Endothelial Cells metabolism, Pericardium cytology, Pericardium metabolism
- Abstract
The organization of an integrated coronary vasculature requires the specification of immature endothelial cells (ECs) into arterial and venous fates based on their localization within the heart. It remains unclear how spatial information controls EC identity and behavior. Here we use single-cell RNA sequencing at key developmental timepoints to interrogate cellular contributions to coronary vessel patterning and maturation. We perform transcriptional profiling to define a heterogenous population of epicardium-derived cells (EPDCs) that express unique chemokine signatures. We identify a population of Slit2+ EPDCs that emerge following epithelial-to-mesenchymal transition (EMT), which we term vascular guidepost cells. We show that the expression of guidepost-derived chemokines such as Slit2 are induced in epicardial cells undergoing EMT, while mesothelium-derived chemokines are silenced. We demonstrate that epicardium-specific deletion of myocardin-related transcription factors in mouse embryos disrupts the expression of key guidance cues and alters EPDC-EC signaling, leading to the persistence of an immature angiogenic EC identity and inappropriate accumulation of ECs on the epicardial surface. Our study suggests that EC pathfinding and fate specification is controlled by a common mechanism and guided by paracrine signaling from EPDCs linking epicardial EMT to EC localization and fate specification in the developing heart.
- Published
- 2021
- Full Text
- View/download PDF
12. Prevention of Fibrosis and Pathological Cardiac Remodeling by Salinomycin.
- Author
-
Burke RM, Dirkx RA Jr, Quijada P, Lighthouse JK, Mohan A, O'Brien M, Wojciechowski W, Woeller CF, Phipps RP, Alexis JD, Ashton JM, and Small EM
- Subjects
- Angiotensin II pharmacology, Animals, Cell Survival drug effects, Disease Models, Animal, Fibrosis, Gene Expression, Heart Failure drug therapy, Heart Failure pathology, Humans, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction pathology, NIH 3T3 Cells, Pyrans isolation & purification, Ventricular Remodeling drug effects, p38 Mitogen-Activated Protein Kinases drug effects, p38 Mitogen-Activated Protein Kinases metabolism, rho-Associated Kinases drug effects, rho-Associated Kinases metabolism, Antifibrotic Agents pharmacology, Cardiomegaly prevention & control, Extracellular Matrix, Myocardium pathology, Myofibroblasts drug effects, Pyrans pharmacology
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
13. Sarcomeres regulate murine cardiomyocyte maturation through MRTF-SRF signaling.
- Author
-
Guo Y, Cao Y, Jardin BD, Sethi I, Ma Q, Moghadaszadeh B, Troiano EC, Mazumdar N, Trembley MA, Small EM, Yuan GC, Beggs AH, and Pu WT
- Subjects
- Actinin metabolism, Animals, Cell Nucleus metabolism, Cytoskeleton metabolism, Gene Expression Regulation genetics, Mice, Mitochondria metabolism, Morphogenesis, Mutation, Myocytes, Cardiac pathology, Sarcomeres pathology, Serum Response Factor metabolism, Signal Transduction, Trans-Activators metabolism, Transcription Factors metabolism, Actinin genetics, Myocytes, Cardiac metabolism, Sarcomeres metabolism
- Abstract
The paucity of knowledge about cardiomyocyte maturation is a major bottleneck in cardiac regenerative medicine. In development, cardiomyocyte maturation is characterized by orchestrated structural, transcriptional, and functional specializations that occur mainly at the perinatal stage. Sarcomeres are the key cytoskeletal structures that regulate the ultrastructural maturation of other organelles, but whether sarcomeres modulate the signal transduction pathways that are essential for cardiomyocyte maturation remains unclear. To address this question, here we generated mice with cardiomyocyte-specific, mosaic, and hypomorphic mutations of α-actinin-2 ( Actn2 ) to study the cell-autonomous roles of sarcomeres in postnatal cardiomyocyte maturation. Actn2 mutation resulted in defective structural maturation of transverse-tubules and mitochondria. In addition, Actn2 mutation triggered transcriptional dysregulation, including abnormal expression of key sarcomeric and mitochondrial genes, and profound impairment of the normal progression of maturational gene expression. Mechanistically, the transcriptional changes in Actn2 mutant cardiomyocytes strongly correlated with those in cardiomyocytes deleted of serum response factor (SRF), a critical transcription factor that regulates cardiomyocyte maturation. Actn2 mutation increased the monomeric form of cardiac α-actin, which interacted with the SRF cofactor MRTFA and perturbed its nuclear localization. Overexpression of a dominant-negative MRTFA mutant was sufficient to recapitulate the morphological and transcriptional defects in Actn2 and Srf mutant cardiomyocytes. Together, these data indicate that Actn2 -based sarcomere organization regulates structural and transcriptional maturation of cardiomyocytes through MRTF-SRF signaling., Competing Interests: The authors declare no conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
14. Fibroblast contributions to ischemic cardiac remodeling.
- Author
-
Burke RM, Burgos Villar KN, and Small EM
- Subjects
- Animals, Extracellular Matrix metabolism, Fibroblasts cytology, Humans, Matrix Metalloproteinases metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardium cytology, Myocardium metabolism, Myocardium pathology, Proteoglycans metabolism, Transforming Growth Factor beta1 metabolism, Fibroblasts metabolism, Ventricular Remodeling
- Published
- 2021
- Full Text
- View/download PDF
15. Cut the YAP: Limiting Fibrosis in Pathologic Cardiac Remodeling.
- Author
-
Small EM and Brooks AC
- Published
- 2020
- Full Text
- View/download PDF
16. Regulation of UV damage repair in quiescent yeast cells.
- Author
-
Long LJ, Lee PH, Small EM, Hillyer C, Guo Y, and Osley MA
- Subjects
- Cell Cycle, DNA, Fungal metabolism, DNA, Fungal radiation effects, DNA-Binding Proteins metabolism, DNA-Directed DNA Polymerase metabolism, Mutagenesis, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae radiation effects, Saccharomyces cerevisiae Proteins metabolism, DNA Damage, DNA Repair, Saccharomyces cerevisiae genetics, Ultraviolet Rays
- Abstract
Non-growing quiescent cells face special challenges when repairing lesions produced by exogenous DNA damaging agents. These challenges include the global repression of transcription and translation and a compacted chromatin structure. We investigated how quiescent yeast cells regulated the repair of DNA lesions produced by UV irradiation. We found that UV lesions were excised and repaired in quiescent cells before their re-entry into S phase, and that lesion repair was correlated with high levels of Rad7, a recognition factor in the global genome repair sub-pathway of nucleotide excision repair (GGR-NER). UV exposure led to an increased frequency of mutations that included C->T transitions and T > A transversions. Mutagenesis was dependent on the error-prone translesion synthesis (TLS) DNA polymerase, Pol zeta, which was the only DNA polymerase present in detectable levels in quiescent cells. Across the genome of quiescent cells, UV-induced mutations showed an association with exons that contained H3K36 or H3K79 trimethylation but not with those bound by RNA polymerase II. Together, the data suggest that the distinct physiological state and chromatin structure of quiescent cells contribute to its regulation of UV damage repair., Competing Interests: Declaration of Competing Interest None of the authors have any conflict of interests to report., (Copyright © 2020 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. The Role of the Epicardium During Heart Development and Repair.
- Author
-
Quijada P, Trembley MA, and Small EM
- Subjects
- Animals, Humans, Myocardium cytology, Myocardium metabolism, Paracrine Communication, Pericardium cytology, Pericardium metabolism, Pericardium physiology, Heart Diseases etiology, Pericardium growth & development, Regeneration
- Abstract
The heart is lined by a single layer of mesothelial cells called the epicardium that provides important cellular contributions for embryonic heart formation. The epicardium harbors a population of progenitor cells that undergo epithelial-to-mesenchymal transition displaying characteristic conversion of planar epithelial cells into multipolar and invasive mesenchymal cells before differentiating into nonmyocyte cardiac lineages, such as vascular smooth muscle cells, pericytes, and fibroblasts. The epicardium is also a source of paracrine cues that are essential for fetal cardiac growth, coronary vessel patterning, and regenerative heart repair. Although the epicardium becomes dormant after birth, cardiac injury reactivates developmental gene programs that stimulate epithelial-to-mesenchymal transition; however, it is not clear how the epicardium contributes to disease progression or repair in the adult. In this review, we will summarize the molecular mechanisms that control epicardium-derived progenitor cell migration, and the functional contributions of the epicardium to heart formation and cardiomyopathy. Future perspectives will be presented to highlight emerging therapeutic strategies aimed at harnessing the regenerative potential of the fetal epicardium for cardiac repair.
- Published
- 2020
- Full Text
- View/download PDF
18. A Novel Role of Cyclic Nucleotide Phosphodiesterase 10A in Pathological Cardiac Remodeling and Dysfunction.
- Author
-
Chen S, Zhang Y, Lighthouse JK, Mickelsen DM, Wu J, Yao P, Small EM, and Yan C
- Subjects
- Animals, Cardiomegaly genetics, Cardiomegaly pathology, Disease Models, Animal, Fibroblasts pathology, Mice, Mice, Knockout, Myocytes, Cardiac pathology, Phosphoric Diester Hydrolases genetics, Transcriptome, Cardiomegaly enzymology, Fibroblasts enzymology, Myocytes, Cardiac enzymology, Phosphoric Diester Hydrolases metabolism, Ventricular Remodeling
- Abstract
Background: Heart failure is a leading cause of death worldwide. Cyclic nucleotide phosphodiesterases (PDEs), through degradation of cyclic nucleotides, play critical roles in cardiovascular biology and disease. Our preliminary screening studies have revealed PDE10A upregulation in the diseased heart. However, the roles of PDE10A in cardiovascular biology and disease are largely uncharacterized. The current study is aimed to investigate the regulation and function of PDE10A in cardiac cells and in the progression of cardiac remodeling and dysfunction., Methods: We used isolated adult mouse cardiac myocytes and fibroblasts, as well as preclinical mouse models of hypertrophy and heart failure. The PDE10A selective inhibitor TP-10, and global PDE10A knock out mice were used., Results: We found that PDE10A expression remains relatively low in normal and exercised heart tissues. However, PDE10A is significantly upregulated in mouse and human failing hearts. In vitro, PDE10A deficiency or inhibiting PDE10A with selective inhibitor TP-10, attenuated cardiac myocyte pathological hypertrophy induced by Angiotensin II, phenylephrine, and isoproterenol, but did not affect cardiac myocyte physiological hypertrophy induced by IGF-1 (insulin-like growth factor 1). TP-10 also reduced TGF-β (transforming growth factor-β)-stimulated cardiac fibroblast activation, proliferation, migration and extracellular matrix synthesis. TP-10 treatment elevated both cAMP and cGMP levels in cardiac myocytes and cardiac fibroblasts, consistent with PDE10A as a cAMP/cGMP dual-specific PDE. In vivo, global PDE10A deficiency significantly attenuated myocardial hypertrophy, cardiac fibrosis, and dysfunction induced by chronic pressure overload via transverse aorta constriction or chronic neurohormonal stimulation via Angiotensin II infusion. Importantly, we demonstrated that the pharmacological effect of TP-10 is specifically through PDE10A inhibition. In addition, TP-10 is able to reverse pre-established cardiac hypertrophy and dysfunction. RNA-Sequencing and bioinformatics analysis further identified a PDE10A-regualted transcriptome involved in cardiac hypertrophy, fibrosis, and cardiomyopathy., Conclusions: Taken together, our study elucidates a novel role for PDE10A in the regulation of pathological cardiac remodeling and development of heart failure. Given that PDE10A has been proven to be a safe drug target, PDE10A inhibition may represent a novel therapeutic strategy for preventing and treating cardiac diseases associated with cardiac remodeling.
- Published
- 2020
- Full Text
- View/download PDF
19. SPOCK, an R based package for high-throughput analysis of growth rate, survival, and chronological lifespan in yeast.
- Author
-
Small EM, Felker DP, Heath OC, Cantergiani RJ, Osley MA, and McCormick MA
- Abstract
Plate reader-based methods for high-throughput measurement of growth rate, cellular survival, and chronological lifespan are a compelling addition to the already powerful toolbox of budding yeast Saccharomyces cerevisiae genetics. These methods have overcome many of the limits of traditional yeast biology techniques, but also present a new bottleneck at the point of data-analysis. Herein, we describe SPOCK (Survival Percentage and Outgrowth Collection Kit), an R-based package for the analysis of data created by high-throughput plate reader based methods. This package allows for the determination of chronological lifespan, cellular growth rate, and survival in an efficient, robust, and reproducible fashion., Competing Interests: DECLARATION OF COMPETING INTERESTS The authors do not have any conflicts of interest to declare.
- Published
- 2020
- Full Text
- View/download PDF
20. Myofibroblast-specific YY1 promotes liver fibrosis.
- Author
-
Liu H, Zhang S, Xu S, Koroleva M, Small EM, and Jin ZG
- Subjects
- Animals, Cell Line, Gene Deletion, Humans, Liver Cirrhosis genetics, Mice, Mice, Inbred C57BL, Myofibroblasts metabolism, RNA Interference, Liver Cirrhosis pathology, Myofibroblasts pathology, YY1 Transcription Factor genetics
- Abstract
Liver fibrosis is a common consequence of various chronic hepatitis and liver injuries. The myofibroblasts, through the accumulation of extracellular matrix (ECM) proteins, are closely associated with the progression of liver fibrosis. However, the molecular mechanisms underlying transcriptional regulation of fibrogenic genes and ECM proteins in myofibroblasts remain largely unknown. Using tamoxifen inducible myofibroblast-specific Cre-expressing mouse lines with selective deletion of the transcription factor Yin Yang 1 (YY1), here we show that YY1 deletion in myofibroblasts mitigates carbon tetrachloride-induced liver fibrosis. This protective effect of YY1 ablation on liver fibrosis was accompanied with reduced expression of profibrogenic genes and ECM proteins, including TNF-α, TGF-β, PDGF, IL-6, α-SMA and Col1α1 in liver tissues from YY1 mutant mice. Moreover, using the human hepatic stellate cell (HSC) line LX-2, we found that knockdown of YY1 in myofibroblasts by siRNA treatment diminished myofibroblast proliferation, α-SMA expression, and collagen deposition. Collectively, our findings reveal a specific role of YY1 in hepatic myofibroblasts and suggest a new therapeutic strategy for hepatic fibrosis-associated liver diseases., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. Pre-existing fibroblasts of epicardial origin are the primary source of pathological fibrosis in cardiac ischemia and aging.
- Author
-
Quijada P, Misra A, Velasquez LS, Burke RM, Lighthouse JK, Mickelsen DM, Dirkx RA Jr, and Small EM
- Subjects
- Animals, Cell Lineage, Diastole, Fibrosis, Heart physiopathology, Mice, Knockout, Myocardial Ischemia physiopathology, Serum Response Factor metabolism, Stem Cells metabolism, Trans-Activators metabolism, Ventricular Remodeling, WT1 Proteins metabolism, Aging pathology, Fibroblasts pathology, Myocardial Ischemia pathology, Pericardium pathology
- Abstract
Serum response factor (SRF) and the SRF co-activators myocardin-related transcription factors (MRTFs) are essential for epicardium-derived progenitor cell (EPDC)-mobilization during heart development; however, the impact of developmental EPDC deficiencies on adult cardiac physiology has not been evaluated. Here, we utilize the Wilms Tumor-1 (Wt1)-Cre to delete Mrtfs or Srf in the epicardium, which reduced the number of EPDCs in the adult cardiac interstitium. Deficiencies in Wt1-lineage EPDCs prevented the development of cardiac fibrosis and diastolic dysfunction in aged mice. Mice lacking MRTF or SRF in EPDCs also displayed preservation of cardiac function following myocardial infarction partially due to the depletion of Wt1 lineage-derived cells in the infarct. Interestingly, depletion of Wt1-lineage EPDCs allows for the population of the infarct with a Wt1-negative cell lineage with a reduced fibrotic profile. Taken together, our study conclusively demonstrates the contribution of EPDCs to both ischemic cardiac remodeling and the development of diastolic dysfunction in old age, and reveals the existence of an alternative Wt1-negative source of resident fibroblasts that can populate the infarct., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
22. Sacubitril/Valsartan Decreases Cardiac Fibrosis in Left Ventricle Pressure Overload by Restoring PKG Signaling in Cardiac Fibroblasts.
- Author
-
Burke RM, Lighthouse JK, Mickelsen DM, and Small EM
- Subjects
- Angiotensin Receptor Antagonists therapeutic use, Animals, Biphenyl Compounds, Drug Combinations, Fibroblasts metabolism, Fibrosis drug therapy, Heart drug effects, Heart physiopathology, Heart Failure physiopathology, Heart Ventricles physiopathology, Male, Mice, Inbred C57BL, Neprilysin antagonists & inhibitors, Valsartan, Aminobutyrates pharmacology, Cyclic GMP-Dependent Protein Kinases drug effects, Fibroblasts drug effects, Heart Failure drug therapy, Heart Ventricles drug effects, Tetrazoles pharmacology
- Abstract
Background Heart failure (HF) is invariably accompanied by development of cardiac fibrosis, a form of scarring that increases muscular tissue rigidity and decreases cardiac contractility. Cardiac fibrosis arises from a pathological attempt to repair tissue damaged during maladaptive remodeling. Treatment options to block or reverse fibrosis have proven elusive. Neprilysin is an endopeptidase that degrades vasoactive peptides, including atrial natriuretic peptide. Thus, neprilysin inhibition reduces hypertension, ultimately limiting maladaptive cardiac remodeling. LCZ696, which consists of an angiotensin receptor blocker (valsartan [VAL]) and a neprilysin inhibitor (sacubitril [SAC]), was shown to be well tolerated and significantly reduced the risk of death and hospitalization in HF patients with reduced ejection fraction. We hypothesized that SAC/VAL directly inhibits fibroblast activation and development of pathological fibrosis. Methods and Results We used a mouse model of left ventricle pressure overload coupled to in vitro studies in primary mouse and human cardiac fibroblasts (CFs) to study the impact of SAC/VAL on CF activation and cardiac fibrosis. SAC/VAL significantly ameliorated pressure overload-induced cardiac fibrosis by blocking CF activation and proliferation, leading to functional improvement. Mechanistically, the beneficial impact of SAC/VAL at least partially stemmed from restoration of PKG (protein kinase G) signaling in HF patient-derived CF, which inhibited Rho activation associated with myofibroblast transition. Conclusions This study reveals that SAC/VAL acts directly on CF to prevent maladaptive cardiac fibrosis and dysfunction during pressure overload-induced hypertrophy and suggests that SAC/VAL should be evaluated as a direct antifibrotic therapeutic for conditions such as HF with preserved ejection fraction.
- Published
- 2019
- Full Text
- View/download PDF
23. Platelet-derived β2M regulates monocyte inflammatory responses.
- Author
-
Hilt ZT, Pariser DN, Ture SK, Mohan A, Quijada P, Asante AA, Cameron SJ, Sterling JA, Merkel AR, Johanson AL, Jenkins JL, Small EM, McGrath KE, Palis J, Elliott MR, and Morrell CN
- Subjects
- Animals, Cell Differentiation, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Chaperones, Platelet Activation, Receptor, Transforming Growth Factor-beta Type II genetics, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, THP-1 Cells, beta 2-Microglobulin genetics, Blood Platelets metabolism, Monocytes metabolism, beta 2-Microglobulin metabolism
- Abstract
β-2 Microglobulin (β2M) is a molecular chaperone for the major histocompatibility class I (MHC I) complex, hemochromatosis factor protein (HFE), and the neonatal Fc receptor (FcRn), but β2M may also have less understood chaperone-independent functions. Elevated plasma β2M has a direct role in neurocognitive decline and is a risk factor for adverse cardiovascular events. β2M mRNA is present in platelets at very high levels, and β2M is part of the activated platelet releasate. In addition to their more well-studied thrombotic functions, platelets are important immune regulatory cells that release inflammatory molecules and contribute to leukocyte trafficking, activation, and differentiation. We have now found that platelet-derived β2M is a mediator of monocyte proinflammatory differentiation through noncanonical TGFβ receptor signaling. Circulating monocytes from mice lacking β2M only in platelets (Plt-β2M-/-) had a more proreparative monocyte phenotype, in part dependent on increased platelet-derived TGFβ signaling in the absence of β2M. Using a mouse myocardial infarction (MI) model, Plt-β2M-/- mice had limited post-MI proinflammatory monocyte responses and, instead, demonstrated early proreparative monocyte differentiation, profibrotic myofibroblast responses, and a rapid decline in heart function compared with WT mice. These data demonstrate a potentially novel chaperone-independent, monocyte phenotype-regulatory function for platelet β2M and that platelet-derived 2M and TGFβ have opposing roles in monocyte differentiation that may be important in tissue injury responses.
- Published
- 2019
- Full Text
- View/download PDF
24. Activated Human Lung Fibroblasts Produce Extracellular Vesicles with Antifibrotic Prostaglandins.
- Author
-
Lacy SH, Woeller CF, Thatcher TH, Pollock SJ, Small EM, Sime PJ, and Phipps RP
- Subjects
- Cell Differentiation drug effects, Cells, Cultured, Dinoprostone metabolism, Exosomes drug effects, Exosomes metabolism, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Extracellular Vesicles metabolism, Female, Fibroblasts metabolism, Humans, Interleukin-1beta metabolism, Lung drug effects, Lung metabolism, Male, Myofibroblasts drug effects, Myofibroblasts metabolism, Pulmonary Fibrosis drug therapy, Pulmonary Fibrosis metabolism, Signal Transduction drug effects, Transforming Growth Factor beta metabolism, Antifibrinolytic Agents pharmacology, Extracellular Vesicles drug effects, Fibroblasts drug effects, Prostaglandins pharmacology
- Abstract
The differentiation of interstitial lung fibroblasts into contractile myofibroblasts that proliferate and secrete excessive extracellular matrix is critical for the pathogenesis of pulmonary fibrosis. Certain lipid signaling molecules, such as prostaglandins (PGs), can inhibit myofibroblast differentiation. However, the sources and delivery mechanisms of endogenous PGs are undefined. Activated primary human lung fibroblasts (HLFs) produce PGs such as PGE
2 . We report that activation of primary HLFs with IL-1β inhibited transforming growth factor β-induced myofibroblast differentiation in both the IL-1β-treated cells themselves (autocrine signal) and adjacent naive HLFs in cocultures (paracrine signal). Additionally, we demonstrate for the first time that at least some of the antifibrotic effect of activated fibroblasts on nearby naive fibroblasts is carried by exosomes and other extracellular vesicles that contain several PGs, including high levels of the antifibrotic PGE2 . Thus, activated fibroblasts communicate with surrounding cells to limit myofibroblast differentiation and maintain homeostasis. This work opens the way for future research into extracellular vesicle-mediated intercellular signaling in the lung and may inform the development of novel therapies for fibrotic lung diseases.- Published
- 2019
- Full Text
- View/download PDF
25. Exercise promotes a cardioprotective gene program in resident cardiac fibroblasts.
- Author
-
Lighthouse JK, Burke RM, Velasquez LS, Dirkx RA Jr, Aiezza A 2nd, Moravec CS, Alexis JD, Rosenberg A, and Small EM
- Abstract
Exercise and heart disease both induce cardiac remodeling, but only disease causes fibrosis and compromises heart function. The cardioprotective benefits of exercise have been attributed to changes in cardiomyocyte physiology, but the impact of exercise on cardiac fibroblasts (CFs) is unknown. Here, RNA-sequencing reveals rapid divergence of CF transcriptional programs during exercise and disease. Among the differentially expressed programs, NRF2-dependent antioxidant genes - including metallothioneins (Mt1 and Mt2) - are induced in CFs during exercise and suppressed by TGF-β/p38 signaling in disease. In vivo, mice lacking Mt1/2 exhibit signs of cardiac dysfunction in exercise, including cardiac fibrosis, vascular rarefaction, and functional decline. Mechanistically, exogenous MTs derived from fibroblasts are taken up by cultured cardiomyocytes, reducing oxidative damage-dependent cell death. Importantly, suppression of MT expression is conserved in human heart failure. Taken together, this study defines the acute transcriptional response of CFs to exercise and disease and reveals a cardioprotective mechanism that is lost in disease.
- Published
- 2019
- Full Text
- View/download PDF
26. LAMP-2B regulates human cardiomyocyte function by mediating autophagosome-lysosome fusion.
- Author
-
Chi C, Leonard A, Knight WE, Beussman KM, Zhao Y, Cao Y, Londono P, Aune E, Trembley MA, Small EM, Jeong MY, Walker LA, Xu H, Sniadecki NJ, Taylor MR, Buttrick PM, and Song K
- Subjects
- Adaptor Proteins, Vesicular Transport genetics, Adaptor Proteins, Vesicular Transport metabolism, Autophagosomes pathology, Autophagy-Related Proteins genetics, Autophagy-Related Proteins metabolism, Gene Knockout Techniques, Glycogen Storage Disease Type IIb genetics, Glycogen Storage Disease Type IIb pathology, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Lysosomal-Associated Membrane Protein 2 genetics, Lysosomes genetics, Lysosomes pathology, Myocytes, Cardiac pathology, Qa-SNARE Proteins genetics, Qa-SNARE Proteins metabolism, R-SNARE Proteins genetics, R-SNARE Proteins metabolism, Autophagosomes metabolism, Glycogen Storage Disease Type IIb metabolism, Lysosomal-Associated Membrane Protein 2 metabolism, Lysosomes metabolism, Membrane Fusion, Myocytes, Cardiac metabolism
- Abstract
Mutations in lysosomal-associated membrane protein 2 ( LAMP-2 ) gene are associated with Danon disease, which often leads to cardiomyopathy/heart failure through poorly defined mechanisms. Here, we identify the LAMP-2 isoform B (LAMP-2B) as required for autophagosome-lysosome fusion in human cardiomyocytes (CMs). Remarkably, LAMP-2B functions independently of syntaxin 17 (STX17), a protein that is essential for autophagosome-lysosome fusion in non-CMs. Instead, LAMP-2B interacts with autophagy related 14 (ATG14) and vesicle-associated membrane protein 8 (VAMP8) through its C-terminal coiled coil domain (CCD) to promote autophagic fusion. CMs derived from induced pluripotent stem cells (hiPSC-CMs) from Danon patients exhibit decreased colocalization between ATG14 and VAMP8, profound defects in autophagic fusion, as well as mitochondrial and contractile abnormalities. This phenotype was recapitulated by LAMP-2B knockout in non-Danon hiPSC-CMs. Finally, gene correction of LAMP-2 mutation rescues the Danon phenotype. These findings reveal a STX17-independent autophagic fusion mechanism in human CMs, providing an explanation for cardiomyopathy in Danon patients and a foundation for targeting defective LAMP-2B-mediated autophagy to treat this patient population., Competing Interests: Conflict of interest statement: E.M.S. is the recipient of a research grant from Novartis Pharmaceuticals that is not related to the present study.
- Published
- 2019
- Full Text
- View/download PDF
27. Mechanosensitive Gene Regulation by Myocardin-Related Transcription Factors Is Required for Cardiomyocyte Integrity in Load-Induced Ventricular Hypertrophy.
- Author
-
Trembley MA, Quijada P, Agullo-Pascual E, Tylock KM, Colpan M, Dirkx RA Jr, Myers JR, Mickelsen DM, de Mesy Bentley K, Rothenberg E, Moravec CS, Alexis JD, Gregorio CC, Dirksen RT, Delmar M, and Small EM
- Subjects
- Aged, Animals, Animals, Newborn, COS Cells, Case-Control Studies, Chlorocebus aethiops, Connexin 43 genetics, Connexin 43 metabolism, Female, Gene Expression Regulation, Heart Failure genetics, Heart Failure pathology, Heart Failure physiopathology, Humans, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular physiopathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Confocal, Microscopy, Electron, Transmission, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Middle Aged, Myocytes, Cardiac ultrastructure, NIH 3T3 Cells, Single Molecule Imaging, Trans-Activators deficiency, Trans-Activators genetics, Transcription Factors deficiency, Transcription Factors genetics, Ventricular Function, Left, Ventricular Remodeling, Heart Failure metabolism, Hypertrophy, Left Ventricular metabolism, Mechanotransduction, Cellular, Myocytes, Cardiac metabolism, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
Background: Hypertrophic cardiomyocyte growth and dysfunction accompany various forms of heart disease. The mechanisms responsible for transcriptional changes that affect cardiac physiology and the transition to heart failure are not well understood. The intercalated disc (ID) is a specialized intercellular junction coupling cardiomyocyte force transmission and propagation of electrical activity. The ID is gaining attention as a mechanosensitive signaling hub and hotspot for causative mutations in cardiomyopathy., Methods: Transmission electron microscopy, confocal microscopy, and single-molecule localization microscopy were used to examine changes in ID structure and protein localization in the murine and human heart. We conducted detailed cardiac functional assessment and transcriptional profiling of mice lacking myocardin-related transcription factor (MRTF)-A and MRTF-B specifically in adult cardiomyocytes to evaluate the role of mechanosensitive regulation of gene expression in load-induced ventricular remodeling., Results: We found that MRTFs localize to IDs in the healthy human heart and accumulate in the nucleus in heart failure. Although mice lacking MRTFs in adult cardiomyocytes display normal cardiac physiology at baseline, pressure overload leads to rapid heart failure characterized by sarcomere disarray, ID disintegration, chamber dilation and wall thinning, cardiac functional decline, and partially penetrant acute lethality. Transcriptional profiling reveals a program of actin cytoskeleton and cardiomyocyte adhesion genes driven by MRTFs during pressure overload. Indeed, conspicuous remodeling of gap junctions at IDs identified by single-molecule localization microscopy may partially stem from a reduction in Mapre1 expression, which we show is a direct mechanosensitive MRTF target., Conclusions: Our study describes a novel paradigm in which MRTFs control an acute mechanosensitive signaling circuit that coordinates cross-talk between the actin and microtubule cytoskeleton and maintains ID integrity and cardiomyocyte homeostasis in heart disease.
- Published
- 2018
- Full Text
- View/download PDF
28. Small proline-rich protein 2B drives stress-dependent p53 degradation and fibroblast proliferation in heart failure.
- Author
-
Burke RM, Lighthouse JK, Quijada P, Dirkx RA Jr, Rosenberg A, Moravec CS, Alexis JD, and Small EM
- Subjects
- Adult, Aged, Animals, Cornified Envelope Proline-Rich Proteins genetics, Heart Failure genetics, Heart Failure physiopathology, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Myocardium metabolism, Proteolysis, Transforming Growth Factor beta1 genetics, Transforming Growth Factor beta1 metabolism, Tumor Suppressor Protein p53 genetics, Cell Proliferation, Cornified Envelope Proline-Rich Proteins metabolism, Fibroblasts metabolism, Heart Failure metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Heart disease is associated with the accumulation of resident cardiac fibroblasts (CFs) that secrete extracellular matrix (ECM), leading to the development of pathological fibrosis and heart failure. However, the mechanisms underlying resident CF proliferation remain poorly defined. Here, we report that small proline-rich protein 2b ( Sprr2b ) is among the most up-regulated genes in CFs during heart disease. We demonstrate that SPRR2B is a regulatory subunit of the USP7/MDM2-containing ubiquitination complex. SPRR2B stimulates the accumulation of MDM2 and the degradation of p53, thus facilitating the proliferation of pathological CFs. Furthermore, SPRR2B phosphorylation by nonreceptor tyrosine kinases in response to TGF-β1 signaling and free-radical production potentiates SPRR2B activity and cell cycle progression. Knockdown of the Sprr2b gene or inhibition of SPRR2B phosphorylation attenuates USP7/MDM2 binding and p53 degradation, leading to CF cell cycle arrest. Importantly, SPRR2B expression is elevated in cardiac tissue from human heart failure patients and correlates with the proliferative state of patient-derived CFs in a process that is reversed by insulin growth factor-1 signaling. These data establish SPRR2B as a unique component of the USP7/MDM2 ubiquitination complex that drives p53 degradation, CF accumulation, and the development of pathological cardiac fibrosis., Competing Interests: Conflict of interest statement: E.M.S. is the recipient of a research grant from Novartis Pharmaceuticals.
- Published
- 2018
- Full Text
- View/download PDF
29. Serum Response Factor Is Essential for Maintenance of Podocyte Structure and Function.
- Author
-
Guo B, Lyu Q, Slivano OJ, Dirkx R, Christie CK, Czyzyk J, Hezel AF, Gharavi AG, Small EM, and Miano JM
- Subjects
- Actinin genetics, Actins genetics, Animals, Cytoskeleton, Dilatation, Pathologic genetics, Female, Humans, Kidney Tubules, Distal pathology, Kidney Tubules, Proximal pathology, Male, Mice, Mice, Knockout, Podocytes physiology, RNA, Messenger metabolism, Receptor, Fibroblast Growth Factor, Type 1 genetics, Repressor Proteins genetics, Serum Response Factor genetics, WT1 Proteins, Actins metabolism, Podocytes metabolism, Podocytes ultrastructure, Serum Response Factor physiology, Trans-Activators genetics, Transcription Factors genetics
- Abstract
Podocytes contain an intricate actin cytoskeleton that is essential for the specialized function of this cell type in renal filtration. Serum response factor (SRF) is a master transcription factor for the actin cytoskeleton, but the in vivo expression and function of SRF in podocytes are unknown. We found that SRF protein colocalizes with podocyte markers in human and mouse kidneys. Compared with littermate controls, mice in which the Srf gene was conditionally inactivated with NPHS2 - Cre exhibited early postnatal proteinuria, hypoalbuminemia, and azotemia. Histologic changes in the mutant mice included glomerular capillary dilation and mild glomerulosclerosis, with reduced expression of multiple canonical podocyte markers. We also noted tubular dilation, cell proliferation, and protein casts as well as reactive changes in mesangial cells and interstitial inflammation. Ultrastructure analysis disclosed foot process effacement with loss of slit diaphragms. To ascertain the importance of SRF cofactors in podocyte function, we disabled the myocardin-related transcription factor A and B genes. Although loss of either SRF cofactor alone had no observable effect in the kidney, deficiency of both recapitulated the Srf -null phenotype. These results establish a vital role for SRF and two SRF cofactors in the maintenance of podocyte structure and function., (Copyright © 2018 by the American Society of Nephrology.)
- Published
- 2018
- Full Text
- View/download PDF
30. Tension Creates an Endoreplication Wavefront that Leads Regeneration of Epicardial Tissue.
- Author
-
Cao J, Wang J, Jackman CP, Cox AH, Trembley MA, Balowski JJ, Cox BD, De Simone A, Dickson AL, Di Talia S, Small EM, Kiehart DP, Bursac N, and Poss KD
- Subjects
- Animals, Biomechanical Phenomena, Cell Movement, Giant Cells pathology, Hypertrophy, Mice, Inbred C57BL, Mitosis, Polyploidy, Zebrafish, Endoreduplication, Pericardium physiology, Regeneration
- Abstract
Mechanisms that control cell-cycle dynamics during tissue regeneration require elucidation. Here we find in zebrafish that regeneration of the epicardium, the mesothelial covering of the heart, is mediated by two phenotypically distinct epicardial cell subpopulations. These include a front of large, multinucleate leader cells, trailed by follower cells that divide to produce small, mononucleate daughters. By using live imaging of cell-cycle dynamics, we show that leader cells form by spatiotemporally regulated endoreplication, caused primarily by cytokinesis failure. Leader cells display greater velocities and mechanical tension within the epicardial tissue sheet, and experimentally induced tension anisotropy stimulates ectopic endoreplication. Unbalancing epicardial cell-cycle dynamics with chemical modulators indicated autonomous regenerative capacity in both leader and follower cells, with leaders displaying an enhanced capacity for surface coverage. Our findings provide evidence that mechanical tension can regulate cell-cycle dynamics in regenerating tissue, stratifying the source cell features to improve repair., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. β-Adrenergic Blockade in Ischemia-Reperfusion Injury: βARKing Up a New Tree.
- Author
-
Small EM and Burke RM
- Subjects
- Adrenergic Agents, Fibroblasts, Humans, Trees, Heart Failure, Myocardial Ischemia, Reperfusion Injury
- Published
- 2017
- Full Text
- View/download PDF
32. PDE1C deficiency antagonizes pathological cardiac remodeling and dysfunction.
- Author
-
Knight WE, Chen S, Zhang Y, Oikawa M, Wu M, Zhou Q, Miller CL, Cai Y, Mickelsen DM, Moravec C, Small EM, Abe J, and Yan C
- Abstract
Cyclic nucleotide phosphodiesterase 1C (PDE1C) represents a major phosphodiesterase activity in human myocardium, but its function in the heart remains unknown. Using genetic and pharmacological approaches, we studied the expression, regulation, function, and underlying mechanisms of PDE1C in the pathogenesis of cardiac remodeling and dysfunction. PDE1C expression is up-regulated in mouse and human failing hearts and is highly expressed in cardiac myocytes but not in fibroblasts. In adult mouse cardiac myocytes, PDE1C deficiency or inhibition attenuated myocyte death and apoptosis, which was largely dependent on cyclic AMP/PKA and PI3K/AKT signaling. PDE1C deficiency also attenuated cardiac myocyte hypertrophy in a PKA-dependent manner. Conditioned medium taken from PDE1C-deficient cardiac myocytes attenuated TGF-β-stimulated cardiac fibroblast activation through a mechanism involving the crosstalk between cardiac myocytes and fibroblasts. In vivo, cardiac remodeling and dysfunction induced by transverse aortic constriction, including myocardial hypertrophy, apoptosis, cardiac fibrosis, and loss of contractile function, were significantly attenuated in PDE1C-knockout mice relative to wild-type mice. These results indicate that PDE1C activation plays a causative role in pathological cardiac remodeling and dysfunction. Given the continued development of highly specific PDE1 inhibitors and the high expression level of PDE1C in the human heart, our findings could have considerable therapeutic significance., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
33. Epicardial Outgrowth Culture Assay and Ex Vivo Assessment of Epicardial-derived Cell Migration.
- Author
-
Trembley MA, Velasquez LS, and Small EM
- Subjects
- Animals, Cell Differentiation physiology, Female, Mice, Mice, Inbred C57BL, Mice, Knockout, Nuclear Proteins metabolism, Organ Culture Techniques, Pregnancy, Trans-Activators metabolism, Transcription Factors metabolism, Cell Movement physiology, Pericardium cytology, Pericardium physiology
- Abstract
A single layer of epicardial cells lines the heart, providing paracrine factors that stimulate cardiomyocyte proliferation and directly contributing cardiovascular progenitors during development and disease. While a number of factors have been implicated in epicardium-derived cell (EPDC) mobilization, the mechanisms governing their subsequent migration and differentiation are poorly understood. Here, we present in vitro and ex vivo strategies to study EPDC motility and differentiation. First, we describe a method of obtaining primary epicardial cells by outgrowth culture from the embryonic mouse heart. We also introduce a detailed protocol to assess three-dimensional migration of labeled EPDC in an organ culture system. We provide evidence using these techniques that genetic deletion of myocardin-related transcription factors in the epicardium attenuates EPDC migration. This approach serves as a platform to evaluate candidate modifiers of EPDC biology and could be used to develop genetic or chemical screens to identify novel regulators of EPDC mobilization that might be useful for cardiac repair.
- Published
- 2016
- Full Text
- View/download PDF
34. Complex Interdependence Regulates Heterotypic Transcription Factor Distribution and Coordinates Cardiogenesis.
- Author
-
Luna-Zurita L, Stirnimann CU, Glatt S, Kaynak BL, Thomas S, Baudin F, Samee MA, He D, Small EM, Mileikovsky M, Nagy A, Holloway AK, Pollard KS, Müller CW, and Bruneau BG
- Subjects
- Animals, Cell Differentiation, Crystallography, X-Ray, Embryo, Mammalian metabolism, Homeobox Protein Nkx-2.5, Homeodomain Proteins genetics, Mice, Mice, Transgenic, Models, Molecular, Myocardium metabolism, Promoter Regions, Genetic, Protein Interaction Domains and Motifs, T-Box Domain Proteins genetics, Transcription Factors genetics, GATA4 Transcription Factor metabolism, Homeodomain Proteins metabolism, Myocardium cytology, Organogenesis, T-Box Domain Proteins metabolism, Transcription Factors metabolism
- Abstract
Transcription factors (TFs) are thought to function with partners to achieve specificity and precise quantitative outputs. In the developing heart, heterotypic TF interactions, such as between the T-box TF TBX5 and the homeodomain TF NKX2-5, have been proposed as a mechanism for human congenital heart defects. We report extensive and complex interdependent genomic occupancy of TBX5, NKX2-5, and the zinc finger TF GATA4 coordinately controlling cardiac gene expression, differentiation, and morphogenesis. Interdependent binding serves not only to co-regulate gene expression but also to prevent TFs from distributing to ectopic loci and activate lineage-inappropriate genes. We define preferential motif arrangements for TBX5 and NKX2-5 cooperative binding sites, supported at the atomic level by their co-crystal structure bound to DNA, revealing a direct interaction between the two factors and induced DNA bending. Complex interdependent binding mechanisms reveal tightly regulated TF genomic distribution and define a combinatorial logic for heterotypic TF regulation of differentiation., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Transcriptional control of cardiac fibroblast plasticity.
- Author
-
Lighthouse JK and Small EM
- Subjects
- Animals, Calcineurin genetics, Calcineurin metabolism, Cell Differentiation, Fibrosis, Humans, Myocardium cytology, Myofibroblasts cytology, NFATC Transcription Factors genetics, NFATC Transcription Factors metabolism, Serum Response Factor genetics, Serum Response Factor metabolism, Signal Transduction, TRPC Cation Channels genetics, TRPC Cation Channels metabolism, Trans-Activators genetics, Trans-Activators metabolism, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Gene Expression Regulation, Myocardium metabolism, Myofibroblasts metabolism, Transcription, Genetic
- Abstract
Cardiac fibroblasts help maintain the normal architecture of the healthy heart and are responsible for scar formation and the healing response to pathological insults. Various genetic, biomechanical, or humoral factors stimulate fibroblasts to become contractile smooth muscle-like cells called myofibroblasts that secrete large amounts of extracellular matrix. Unfortunately, unchecked myofibroblast activation in heart disease leads to pathological fibrosis, which is a major risk factor for the development of cardiac arrhythmias and heart failure. A better understanding of the molecular mechanisms that control fibroblast plasticity and myofibroblast activation is essential to develop novel strategies to specifically target pathological cardiac fibrosis without disrupting the adaptive healing response. This review highlights the major transcriptional mediators of fibroblast origin and function in development and disease. The contribution of the fetal epicardial gene program will be discussed in the context of fibroblast origin in development and following injury, primarily focusing on Tcf21 and C/EBP. We will also highlight the major transcriptional regulatory axes that control fibroblast plasticity in the adult heart, including transforming growth factor β (TGFβ)/Smad signaling, the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) axis, and Calcineurin/transient receptor potential channel (TRP)/nuclear factor of activated T-Cell (NFAT) signaling. Finally, we will discuss recent strategies to divert the fibroblast transcriptional program in an effort to promote cardiomyocyte regeneration. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling"., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels.
- Author
-
Trembley MA, Velasquez LS, de Mesy Bentley KL, and Small EM
- Subjects
- Animals, COS Cells, Cell Differentiation drug effects, Cell Nucleus drug effects, Cell Nucleus metabolism, Chlorocebus aethiops, Coronary Vessels drug effects, Coronary Vessels metabolism, Embryo, Mammalian drug effects, Embryo, Mammalian metabolism, Embryo, Mammalian pathology, Mice, Inbred C57BL, Neovascularization, Physiologic drug effects, Pericardium metabolism, Pericardium ultrastructure, Pericytes cytology, Pericytes drug effects, Serum Response Factor metabolism, Trans-Activators genetics, Transcription Factors genetics, Transforming Growth Factor beta1 pharmacology, Cell Movement drug effects, Coronary Vessels growth & development, Pericardium cytology, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
An important pool of cardiovascular progenitor cells arises from the epicardium, a single layer of mesothelium lining the heart. Epicardium-derived progenitor cell (EPDC) formation requires epithelial-to-mesenchymal transition (EMT) and the subsequent migration of these cells into the sub-epicardial space. Although some of the physiological signals that promote EMT are understood, the functional mediators of EPDC motility and differentiation are not known. Here, we identify a novel regulatory mechanism of EPDC mobilization. Myocardin-related transcription factor (MRTF)-A and MRTF-B (MKL1 and MKL2, respectively) are enriched in the perinuclear space of epicardial cells during development. Transforming growth factor (TGF)-β signaling and disassembly of cell contacts leads to nuclear accumulation of MRTFs and the activation of the motile gene expression program. Conditional ablation of Mrtfa and Mrtfb specifically in the epicardium disrupts cell migration and leads to sub-epicardial hemorrhage, partially stemming from the depletion of coronary pericytes. Using lineage-tracing analyses, we demonstrate that sub-epicardial pericytes arise from EPDCs in a process that requires the MRTF-dependent motile gene expression program. These findings provide novel mechanisms linking EPDC motility and differentiation, shed light on the transcriptional control of coronary microvascular maturation and suggest novel therapeutic strategies to manipulate epicardium-derived progenitor cells for cardiac repair., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
37. Antithrombotic treatment in transcatheter aortic valve implantation: insights for cerebrovascular and bleeding events.
- Author
-
Rodés-Cabau J, Dauerman HL, Cohen MG, Mehran R, Small EM, Smyth SS, Costa MA, Mega JL, O'Donoghue ML, Ohman EM, and Becker RC
- Subjects
- Animals, Cerebrovascular Disorders diagnosis, Cerebrovascular Disorders epidemiology, Humans, Postoperative Hemorrhage diagnosis, Postoperative Hemorrhage epidemiology, Risk Factors, Treatment Outcome, Cardiac Catheterization adverse effects, Cerebrovascular Disorders prevention & control, Fibrinolytic Agents therapeutic use, Heart Valve Prosthesis Implantation adverse effects, Postoperative Hemorrhage prevention & control
- Abstract
Transcatheter aortic valve implantation (TAVI) has emerged as a therapeutic alternative for patients with symptomatic aortic stenosis at high or prohibitive surgical risk. However, patients undergoing TAVI are also at high risk for both bleeding and stroke complications, and specific mechanical aspects of the procedure itself can increase the risk of these complications. The mechanisms of periprocedural bleeding complications seem to relate mainly to vascular/access site complications (related to the use of large catheters in a very old and frail elderly population), whereas the pathophysiology of cerebrovascular events remains largely unknown. Further, although mechanical complications, especially the interaction between the valve prosthesis and the native aortic valve, may play a major role in events that occur during TAVI, post-procedural events might also be related to a prothrombotic environment or state generated by the implanted valve, the occurrence of atrial arrhythmias, and associated comorbidities. Antithrombotic therapy in the setting of TAVI has been empirically determined, and unfractionated heparin during the procedure followed by dual antiplatelet therapy with aspirin (indefinitely) and clopidogrel (1 to 6 months) is the most commonly recommended treatment. However, bleeding and cerebrovascular events are common; these may be modifiable with optimization of periprocedural and post-procedural pharmacology. Further, as the field of antiplatelet and anticoagulant therapy evolves, potential drug combinations will multiply, introducing variability in treatment. Randomized trials are the best path forward to determine the balance between the efficacy and risks of antithrombotic treatment in this high risk-population., (Copyright © 2013 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
38. Activation of MRTF-A-dependent gene expression with a small molecule promotes myofibroblast differentiation and wound healing.
- Author
-
Velasquez LS, Sutherland LB, Liu Z, Grinnell F, Kamm KE, Schneider JW, Olson EN, and Small EM
- Subjects
- Androstenols pharmacology, Animals, Dermis pathology, Mice, Myofibroblasts pathology, Transforming Growth Factor beta1 pharmacology, Cell Differentiation, Dermis injuries, Dermis metabolism, Gene Expression Regulation, Myofibroblasts metabolism, Trans-Activators metabolism, Wound Healing
- Abstract
Myocardin-related transcription factors (MRTFs) regulate cellular contractility and motility by associating with serum response factor (SRF) and activating genes involved in cytoskeletal dynamics. We reported previously that MRTF-A contributes to pathological cardiac remodeling by promoting differentiation of fibroblasts to myofibroblasts following myocardial infarction. Here, we show that forced expression of MRTF-A in dermal fibroblasts stimulates contraction of a collagen matrix, whereas contractility of MRTF-A null fibroblasts is impaired under basal conditions and in response to TGF-β1 stimulation. We also identify an isoxazole ring-containing small molecule, previously shown to induce smooth muscle α-actin gene expression in cardiac progenitor cells, as an agonist of myofibroblast differentiation. Isoxazole stimulates myofibroblast differentiation via induction of MRTF-A-dependent gene expression. The MRTF-SRF signaling axis is activated in response to skin injury, and treatment of dermal wounds with isoxazole accelerates wound closure and suppresses the inflammatory response. These results reveal an important role for MRTF-SRF signaling in dermal myofibroblast differentiation and wound healing and suggest that targeting MRTFs pharmacologically may prove useful in treating diseases associated with inappropriate myofibroblast activity.
- Published
- 2013
- Full Text
- View/download PDF
39. The actin-MRTF-SRF gene regulatory axis and myofibroblast differentiation.
- Author
-
Small EM
- Subjects
- Animals, Fibrosis, Gene Expression Regulation, Heart Diseases genetics, Heart Diseases pathology, Heart Diseases therapy, Humans, Myocardium pathology, Myofibroblasts pathology, Phenotype, Signal Transduction, Actins metabolism, Cell Differentiation genetics, Heart Diseases metabolism, Myocardium metabolism, Myofibroblasts metabolism, Serum Response Factor metabolism, Transcription Factors metabolism
- Abstract
Cardiac fibroblasts are responsible for necrotic tissue replacement and scar formation after myocardial infarction (MI) and contribute to remodeling in response to pathological stimuli. This response to insult or injury is largely due to the phenotypic plasticity of fibroblasts. When fibroblasts encounter environmental disturbances, whether biomechanical or humoral, they often transform into smooth muscle-like, contractile cells called "myofibroblasts." The signals that control myofibroblast differentiation include the transforming growth factor (TGF)-β1-Smad pathway and Rho GTPase-dependent actin polymerization. Recent evidence implicates serum response factor (SRF) and the myocardin-related transcription factors (MRTFs) as key mediators of the contractile gene program in response to TGF-β1 or RhoA signaling. This review highlights the function of myofibroblasts in cardiac remodeling and the role of the actin-MRTF-SRF signaling axis in regulating this process.
- Published
- 2012
- Full Text
- View/download PDF
40. MicroRNA133a: a new variable in vascular smooth muscle cell phenotypic switching.
- Author
-
Miano JM and Small EM
- Subjects
- Animals, Gene Targeting methods, Humans, MicroRNAs genetics, Muscle, Smooth, Vascular physiology, Myocytes, Smooth Muscle physiology, Phenotype
- Published
- 2011
- Full Text
- View/download PDF
41. Pervasive roles of microRNAs in cardiovascular biology.
- Author
-
Small EM and Olson EN
- Subjects
- Animals, Blood Vessels growth & development, Blood Vessels metabolism, Cardiovascular Diseases diagnosis, Cardiovascular System embryology, Cardiovascular System growth & development, Humans, MicroRNAs antagonists & inhibitors, Mutation, Cardiovascular Diseases genetics, Cardiovascular Diseases therapy, Cardiovascular System metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
First recognized as regulators of development in worms and fruitflies, microRNAs are emerging as pivotal modulators of mammalian cardiovascular development and disease. Individual microRNAs modulate the expression of collections of messenger RNA targets that often have related functions, thereby governing complex biological processes. The wideranging functions of microRNAs in the cardiovascular system have provided new perspectives on disease mechanisms and have revealed intriguing therapeutic targets, as well as diagnostics, for a variety of cardiovascular disorders.
- Published
- 2011
- Full Text
- View/download PDF
42. MicroRNA-218 regulates vascular patterning by modulation of Slit-Robo signaling.
- Author
-
Small EM, Sutherland LB, Rajagopalan KN, Wang S, and Olson EN
- Subjects
- Animals, Base Sequence, COS Cells, Cells, Cultured, Chlorocebus aethiops, Gene Expression Regulation, Developmental physiology, Glycoproteins physiology, Heparitin Sulfate antagonists & inhibitors, Heparitin Sulfate biosynthesis, Heparitin Sulfate genetics, Intercellular Signaling Peptides and Proteins genetics, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, MicroRNAs antagonists & inhibitors, MicroRNAs genetics, Molecular Sequence Data, Neovascularization, Physiologic genetics, Nerve Tissue Proteins genetics, Nerve Tissue Proteins physiology, Receptors, Immunologic genetics, Retinal Vessels physiology, Transcription, Genetic, Roundabout Proteins, Glycoproteins antagonists & inhibitors, MicroRNAs physiology, Nerve Tissue Proteins antagonists & inhibitors, Receptors, Immunologic antagonists & inhibitors, Retinal Vessels embryology, Signal Transduction genetics
- Abstract
Rationale: Establishment of a functional vasculature requires the interconnection and remodeling of nascent blood vessels. Precise regulation of factors that influence endothelial cell migration and function is essential for these stereotypical vascular patterning events. The secreted Slit ligands and their Robo receptors constitute a critical signaling pathway controlling the directed migration of both neurons and vascular endothelial cells during embryonic development, but the mechanisms of their regulation are incompletely understood., Objective: To identify microRNAs regulating aspects of the Slit-Robo pathway and vascular patterning., Methods and Results: Here, we provide evidence that microRNA (miR)-218, which is encoded by an intron of the Slit genes, inhibits the expression of Robo1 and Robo2 and multiple components of the heparan sulfate biosynthetic pathway. Using in vitro and in vivo approaches, we demonstrate that miR-218 directly represses the expression of Robo1, Robo2, and glucuronyl C5-epimerase (GLCE), and that an intact miR-218-Slit-Robo regulatory network is essential for normal vascularization of the retina. Knockdown of miR-218 results in aberrant regulation of this signaling axis, abnormal endothelial cell migration, and reduced complexity of the retinal vasculature., Conclusions: Our findings link Slit gene expression to the posttranscriptional regulation of Robo receptors and heparan sulfate biosynthetic enzymes, allowing for precise control over vascular guidance cues influencing the organization of blood vessels during development.
- Published
- 2010
- Full Text
- View/download PDF
43. Myocardin-related transcription factor-a controls myofibroblast activation and fibrosis in response to myocardial infarction.
- Author
-
Small EM, Thatcher JE, Sutherland LB, Kinoshita H, Gerard RD, Richardson JA, Dimaio JM, Sadek H, Kuwahara K, and Olson EN
- Subjects
- Amides pharmacology, Angiotensin II administration & dosage, Animals, Base Sequence, COS Cells, Chlorocebus aethiops, Collagen genetics, Collagen Type I, Disease Models, Animal, Extracellular Matrix Proteins genetics, Fibroblasts drug effects, Fibroblasts pathology, Fibrosis, Male, Mice, Mice, Knockout, Molecular Sequence Data, Myocardial Infarction genetics, Myocardial Infarction pathology, Myocardium pathology, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, Phenotype, Promoter Regions, Genetic, Protein Kinase Inhibitors pharmacology, Pyridines pharmacology, Time Factors, Trans-Activators deficiency, Trans-Activators genetics, Transcription, Genetic, Transfection, Transforming Growth Factor beta1 metabolism, rho-Associated Kinases metabolism, Cell Transdifferentiation drug effects, Cell Transdifferentiation genetics, Extracellular Matrix Proteins metabolism, Fibroblasts metabolism, Myocardial Infarction metabolism, Myocardium metabolism, Myocytes, Smooth Muscle metabolism, Trans-Activators metabolism, Ventricular Remodeling drug effects, Ventricular Remodeling genetics
- Abstract
Rationale: Myocardial infarction (MI) results in loss of cardiac myocytes in the ischemic zone of the heart, followed by fibrosis and scar formation, which diminish cardiac contractility and impede angiogenesis and repair. Myofibroblasts, a specialized cell type that switches from a fibroblast-like state to a contractile, smooth muscle-like state, are believed to be primarily responsible for fibrosis of the injured heart and other tissues, although the transcriptional mediators of fibrosis and myofibroblast activation remain poorly defined. Myocardin-related transcription factors (MRTFs) are serum response factor (SRF) cofactors that promote a smooth muscle phenotype and are emerging as components of stress-responsive signaling., Objective: We aimed to examine the effect of MRTF-A on cardiac remodeling and fibrosis., Methods and Results: Here, we show that MRTF-A controls the expression of a fibrotic gene program that includes genes involved in extracellular matrix production and smooth muscle cell differentiation in the heart. In MRTF-A-null mice, fibrosis and scar formation following MI or angiotensin II treatment are dramatically diminished compared with wild-type littermates. This protective effect of MRTF-A deletion is associated with a reduction in expression of fibrosis-associated genes, including collagen 1a2, a direct transcriptional target of SRF/MRTF-A., Conclusions: We conclude that MRTF-A regulates myofibroblast activation and fibrosis in response to the renin-angiotensin system and post-MI remodeling.
- Published
- 2010
- Full Text
- View/download PDF
44. Regulation of PI3-kinase/Akt signaling by muscle-enriched microRNA-486.
- Author
-
Small EM, O'Rourke JR, Moresi V, Sutherland LB, McAnally J, Gerard RD, Richardson JA, and Olson EN
- Subjects
- Animals, Blotting, Northern, Electrophoretic Mobility Shift Assay, In Situ Hybridization, Mice, Mice, Transgenic, Oligonucleotide Array Sequence Analysis, Rats, Signal Transduction, Trans-Activators metabolism, MicroRNAs metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
microRNAs (miRNAs) play key roles in modulating a variety of cellular processes through repression of mRNA targets. In a screen for miRNAs regulated by myocardin-related transcription factor-A (MRTF-A), a coactivator of serum response factor (SRF), we discovered a muscle-enriched miRNA, miR-486, controlled by an alternative promoter within intron 40 of the Ankyrin-1 gene. Transcription of miR-486 is directly controlled by SRF and MRTF-A, as well as by MyoD. Among the most strongly predicted targets of miR-486 are phosphatase and tensin homolog (PTEN) and Foxo1a, which negatively affect phosphoinositide-3-kinase (PI3K)/Akt signaling. Accordingly, PTEN and Foxo1a protein levels are reduced by miR-486 overexpression, which, in turn, enhances PI3K/Akt signaling. Similarly, we show that MRTF-A promotes PI3K/Akt signaling by up-regulating miR-486 expression. Conversely, inhibition of miR-486 expression enhances the expression of PTEN and Foxo1a and dampens signaling through the PI3K/Akt-signaling pathway. Our findings implicate miR-486 as a downstream mediator of the actions of SRF/MRTF-A and MyoD in muscle cells and as a potential modulator of PI3K/Akt signaling.
- Published
- 2010
- Full Text
- View/download PDF
45. MicroRNAs miR-143 and miR-145 modulate cytoskeletal dynamics and responsiveness of smooth muscle cells to injury.
- Author
-
Xin M, Small EM, Sutherland LB, Qi X, McAnally J, Plato CF, Richardson JA, Bassel-Duby R, and Olson EN
- Subjects
- Actins metabolism, Animals, Base Sequence, Carotid Artery Injuries metabolism, Cells, Cultured, Enhancer Elements, Genetic genetics, Female, Humans, Male, Mice, Mice, Inbred C57BL, MicroRNAs genetics, Molecular Sequence Data, Mutation, Myocytes, Cardiac metabolism, Myocytes, Smooth Muscle pathology, Nuclear Proteins metabolism, Rats, Sequence Alignment, Trans-Activators metabolism, Cytoskeleton metabolism, Gene Expression Regulation, MicroRNAs metabolism, Myocytes, Smooth Muscle metabolism
- Abstract
Vascular injury triggers dedifferentiation and cytoskeletal remodeling of smooth muscle cells (SMCs), culminating in vessel occlusion. Serum response factor (SRF) and its coactivator, myocardin, play a central role in the control of smooth muscle phenotypes by regulating the expression of cytoskeletal genes. We show that SRF and myocardin regulate a cardiovascular-specific microRNA (miRNA) cluster encoding miR-143 and miR-145. To assess the functions of these miRNAs in vivo, we systematically deleted them singly and in combination in mice. Mice lacking both miR-143 and miR-145 are viable and do not display overt abnormalities in smooth muscle differentiation, although they show a significant reduction in blood pressure due to reduced vascular tone. Remarkably, however, neointima formation in response to vascular injury is profoundly impeded in mice lacking these miRNAs, due to disarray of actin stress fibers and diminished migratory activity of SMCs. These abnormalities reflect the regulation of a cadre of modulators of SRF activity and actin dynamics by miR-143 and miR-145. Thus, miR-143 and miR-145 act as integral components of the regulatory network whereby SRF controls cytoskeletal remodeling and phenotypic switching of SMCs during vascular disease.
- Published
- 2009
- Full Text
- View/download PDF
46. The myocardin-related transcription factor, MASTR, cooperates with MyoD to activate skeletal muscle gene expression.
- Author
-
Meadows SM, Warkman AS, Salanga MC, Small EM, and Krieg PA
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Embryo, Nonmammalian metabolism, Genome, Molecular Sequence Data, Muscle, Skeletal metabolism, Myogenic Regulatory Factor 5 metabolism, Transcription Factors genetics, Xenopus, Xenopus Proteins genetics, Gene Expression Regulation, Developmental, Muscle Development genetics, Muscle, Skeletal embryology, MyoD Protein metabolism, Transcription Factors metabolism, Xenopus Proteins metabolism
- Abstract
The myocardin family proteins (myocardin, MRTF-A, and MRTF-B) are serum response factor (SRF) cofactors and potent transcription activators. Gene-ablation studies have indicated important developmental functions for myocardin family proteins primarily in regulation of cardiac and smooth muscle development. Using Xenopus genome and cDNA databases, we identified a myocardin-related transcription factor expressed specifically in the skeletal muscle lineage. Synteny and sequence alignments indicate that this gene is the frog orthologue of mouse MASTR [Creemers EE, Sutherland LB, Oh J, Barbosa AC, Olson EN (2006) Coactivation of MEF2 by the SAP domain proteins myocardin and MASTR. Mol Cell 23:83-96]. Inhibition of MASTR function in the Xenopus embryo by using dominant-negative constructions or morpholino knockdown results in a dramatic reduction in expression of skeletal muscle marker genes. Overexpression of MASTR in whole embryos or embryonic tissue explants induces ectopic expression of muscle marker genes. Furthermore, MASTR cooperates with the myogenic regulatory factors MyoD and Myf5 to activate transcription of skeletal muscle genes. An essential function for MASTR in regulation of myogenic development in the vertebrate embryo has not been previously indicated.
- Published
- 2008
- Full Text
- View/download PDF
47. Essential roles of the bHLH transcription factor Hrt2 in repression of atrial gene expression and maintenance of postnatal cardiac function.
- Author
-
Xin M, Small EM, van Rooij E, Qi X, Richardson JA, Srivastava D, Nakagawa O, and Olson EN
- Subjects
- Animals, Atrial Natriuretic Factor genetics, Female, Heart embryology, Heart Septal Defects, Ventricular etiology, Mice, Rats, Receptors, Notch physiology, Signal Transduction, T-Box Domain Proteins genetics, Basic Helix-Loop-Helix Transcription Factors physiology, Gene Expression Regulation, Heart Atria metabolism, Myocardial Contraction, Repressor Proteins physiology
- Abstract
The basic helix-loop-helix transcriptional repressor Hairy-related transcription factor 2 (Hrt2) is expressed in ventricular, but not atrial, cardiomyocytes, and in endothelial and vascular smooth muscle cells. Mice homozygous for a null mutation of Hrt2 die perinatally from a spectrum of cardiac abnormalities, raising questions about the specific functions of this transcriptional regulator in individual cardiac cell lineages. Using a conditional Hrt2 null allele, we show that cardiomyocyte-specific deletion of Hrt2 in mice results in ectopic activation of atrial genes in ventricular myocardium with an associated impairment of cardiac contractility and a unique distortion in morphology of the right ventricular chamber. Consistent with the atrialization of ventricular gene expression in Hrt2 mutant mice, forced expression of Hrt2 in atrial cardiomyocytes is sufficient to repress atrial cardiac genes. These findings reveal a ventricular myocardial cell-autonomous function for Hrt2 in the suppression of atrial cell identity and the maintenance of postnatal cardiac function.
- Published
- 2007
- Full Text
- View/download PDF
48. The future is now: prospective temporal self-appraisals among defensive pessimists and optimists.
- Author
-
Sanna LJ, Chang EC, Carter SE, and Small EM
- Subjects
- Adaptation, Psychological, Adolescent, Adult, Affect, Female, Humans, Male, Motivation, Personality Inventory, Prospective Studies, Self Concept, Set, Psychology, Character, Defense Mechanisms, Negativism, Self-Assessment
- Abstract
Three studies found that prospective temporal self-appraisals can be part of defensive pessimists' strategy; they felt closer to equally distant negative than positive futures. In Study 1, defensive pessimists felt closer to future failures and reported more negative affect than those considering success. In Study 2, when manipulated negative futures were close, defensive pessimists felt bad and performed well; results suggested that viewing negative futures as close may be part of their natural strategy. Study 3 found that prospective self-appraisals influenced performances through felt preparation. Optimists did not use prospective self-appraisals (Study 1) and their performances were unaffected by manipulated temporal distance (Studies 2 and 3). Discussion centers on prospective self-appraisals and multiple strategies of defensive pessimists.
- Published
- 2006
- Full Text
- View/download PDF
49. Myocardin is sufficient and necessary for cardiac gene expression in Xenopus.
- Author
-
Small EM, Warkman AS, Wang DZ, Sutherland LB, Olson EN, and Krieg PA
- Subjects
- Amino Acid Sequence, Animals, Cardiac Myosins metabolism, Cell Differentiation, Cloning, Molecular, DNA Primers chemistry, DNA-Binding Proteins metabolism, GATA4 Transcription Factor, Gene Expression Regulation, Genetic Markers, Homeobox Protein Nkx-2.5, Homeodomain Proteins metabolism, In Situ Hybridization, Molecular Sequence Data, Myosin Light Chains metabolism, Neurons metabolism, Oligonucleotides, Antisense chemistry, Phenotype, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Amino Acid, T-Box Domain Proteins metabolism, Time Factors, Transcription Factors metabolism, Transcription, Genetic, Transgenes, Xenopus Proteins, Xenopus laevis, Gene Expression Regulation, Developmental, Myocardium metabolism, Nuclear Proteins genetics, Nuclear Proteins physiology, Trans-Activators genetics, Trans-Activators physiology
- Abstract
Myocardin is a cardiac- and smooth muscle-specific cofactor for the ubiquitous transcription factor serum response factor (SRF). Using gain-of-function approaches in the Xenopus embryo, we show that myocardin is sufficient to activate transcription of a wide range of cardiac and smooth muscle differentiation markers in non-muscle cell types. We also demonstrate that, for the myosin light chain 2 gene (MLC2), myocardin cooperates with the zinc-finger transcription factor Gata4 to activate expression. Inhibition of myocardin activity in Xenopus embryos using morpholino knockdown methods results in inhibition of cardiac development and the absence of expression of cardiac differentiation markers and severe disruption of cardiac morphological processes. We conclude that myocardin is an essential component of the regulatory pathway for myocardial differentiation.
- Published
- 2005
- Full Text
- View/download PDF
50. Molecular regulation of cardiac chamber-specific gene expression.
- Author
-
Small EM and Krieg PA
- Subjects
- Animals, Female, Gene Expression Regulation, Developmental, Humans, Male, Molecular Biology, Sensitivity and Specificity, Atrial Natriuretic Factor genetics, Cardiomyopathies genetics, Heart embryology, Myosin Heavy Chains genetics, Myosin Light Chains genetics
- Abstract
This review focuses on recent studies investigating the genetic regulatory mechanisms leading to formation of morphologically, functionally, and molecularly distinct cardiac chambers. The regulation of four representative chamber-specific genes that have been studied in detail is reviewed. These genes include the atrial-specific genes, myosin light chain-2a (MLC2a), slow myosin heavy chain-3 (slow MyHC3), and atrial natriuretic factor (ANF) and the ventricular specific gene, myosin light chain-2v (MLC2v). Comparison of these promoters reveals some generalizations about the regulatory mechanisms involved in chamber-specific gene expression but, equally, indicates the large gaps in the knowledge concerning this intriguing genetic program.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.