Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89.903
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 99-121, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340574

ABSTRACT

B cells are traditionally known for their ability to produce antibodies in the context of adaptive immune responses. However, over the last decade B cells have been increasingly recognized as modulators of both adaptive and innate immune responses, as well as players in an important role in the pathogenesis of a variety of human diseases. Here, after briefly summarizing our current understanding of B cell biology, we present a systematic review of the literature from both animal models and human studies that highlight the important role that B lymphocytes play in cardiac and vascular disease. While many aspects of B cell biology in the vasculature and, to an even greater extent, in the heart remain unclear, B cells are emerging as key regulators of cardiovascular adaptation to injury.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Disease Susceptibility , Adaptive Immunity , Animals , Cardiovascular Diseases/diagnosis , Cytokines/metabolism , Humans , Immunity, Innate , Inflammation Mediators/metabolism
2.
Cell ; 187(17): 4637-4655.e26, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043180

ABSTRACT

The medical burden of stroke extends beyond the brain injury itself and is largely determined by chronic comorbidities that develop secondarily. We hypothesized that these comorbidities might share a common immunological cause, yet chronic effects post-stroke on systemic immunity are underexplored. Here, we identify myeloid innate immune memory as a cause of remote organ dysfunction after stroke. Single-cell sequencing revealed persistent pro-inflammatory changes in monocytes/macrophages in multiple organs up to 3 months after brain injury, notably in the heart, leading to cardiac fibrosis and dysfunction in both mice and stroke patients. IL-1ß was identified as a key driver of epigenetic changes in innate immune memory. These changes could be transplanted to naive mice, inducing cardiac dysfunction. By neutralizing post-stroke IL-1ß or blocking pro-inflammatory monocyte trafficking with a CCR2/5 inhibitor, we prevented post-stroke cardiac dysfunction. Such immune-targeted therapies could potentially prevent various IL-1ß-mediated comorbidities, offering a framework for secondary prevention immunotherapy.


Subject(s)
Brain Injuries , Immunity, Innate , Immunologic Memory , Inflammation , Interleukin-1beta , Mice, Inbred C57BL , Monocytes , Animals , Mice , Interleukin-1beta/metabolism , Brain Injuries/immunology , Humans , Male , Monocytes/metabolism , Monocytes/immunology , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Stroke/complications , Stroke/immunology , Heart Diseases/immunology , Female , Receptors, CCR2/metabolism , Fibrosis , Epigenesis, Genetic , Trained Immunity
3.
Cell ; 186(25): 5587-5605.e27, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38029745

ABSTRACT

The number one cause of human fetal death are defects in heart development. Because the human embryonic heart is inaccessible and the impacts of mutations, drugs, and environmental factors on the specialized functions of different heart compartments are not captured by in vitro models, determining the underlying causes is difficult. Here, we established a human cardioid platform that recapitulates the development of all major embryonic heart compartments, including right and left ventricles, atria, outflow tract, and atrioventricular canal. By leveraging 2D and 3D differentiation, we efficiently generated progenitor subsets with distinct first, anterior, and posterior second heart field identities. This advance enabled the reproducible generation of cardioids with compartment-specific in vivo-like gene expression profiles, morphologies, and functions. We used this platform to unravel the ontogeny of signal and contraction propagation between interacting heart chambers and dissect how mutations, teratogens, and drugs cause compartment-specific defects in the developing human heart.


Subject(s)
Heart Diseases , Heart Ventricles , Heart , Humans , Transcriptome/genetics , Cell Line , Gene Expression Regulation, Developmental , Heart Diseases/genetics , Heart Diseases/metabolism
4.
Cell ; 186(3): 479-496.e23, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736300

ABSTRACT

Using four-dimensional whole-embryo light sheet imaging with improved and accessible computational tools, we longitudinally reconstruct early murine cardiac development at single-cell resolution. Nascent mesoderm progenitors form opposing density and motility gradients, converting the temporal birth sequence of gastrulation into a spatial anterolateral-to-posteromedial arrangement. Migrating precardiac mesoderm does not strictly preserve cellular neighbor relationships, and spatial patterns only become solidified as the cardiac crescent emerges. Progenitors undergo a mesenchymal-to-epithelial transition, with a first heart field (FHF) ridge apposing a motile juxta-cardiac field (JCF). Anchored along the ridge, the FHF epithelium rotates the JCF forward to form the initial heart tube, along with push-pull morphodynamics of the second heart field. In Mesp1 mutants that fail to make a cardiac crescent, mesoderm remains highly motile but directionally incoherent, resulting in density gradient inversion. Our practicable live embryo imaging approach defines spatial origins and behaviors of cardiac progenitors and identifies their unanticipated morphological transitions.


Subject(s)
Heart , Mesoderm , Mice , Animals , Cell Differentiation , Morphogenesis , Embryo, Mammalian , Mammals
5.
Cell ; 184(12): 3299-3317.e22, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34019794

ABSTRACT

Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.


Subject(s)
Heart/embryology , Organogenesis , Organoids/embryology , Activins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Calcium/metabolism , Cell Line , Cell Lineage , Chickens , Endothelial Cells/cytology , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Homeobox Protein Nkx-2.5/metabolism , Humans , Male , Mesoderm/embryology , Models, Biological , Myocardium/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/metabolism
6.
Cell ; 184(20): 5151-5162.e11, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34520724

ABSTRACT

The heartbeat is initiated by voltage-gated sodium channel NaV1.5, which opens rapidly and triggers the cardiac action potential; however, the structural basis for pore opening remains unknown. Here, we blocked fast inactivation with a mutation and captured the elusive open-state structure. The fast inactivation gate moves away from its receptor, allowing asymmetric opening of pore-lining S6 segments, which bend and rotate at their intracellular ends to dilate the activation gate to ∼10 Å diameter. Molecular dynamics analyses predict physiological rates of Na+ conductance. The open-state pore blocker propafenone binds in a high-affinity pose, and drug-access pathways are revealed through the open activation gate and fenestrations. Comparison with mutagenesis results provides a structural map of arrhythmia mutations that target the activation and fast inactivation gates. These results give atomic-level insights into molecular events that underlie generation of the action potential, open-state drug block, and fast inactivation of cardiac sodium channels, which initiate the heartbeat.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Animals , Arrhythmias, Cardiac/genetics , Cryoelectron Microscopy , HEK293 Cells , Heart Rate/drug effects , Humans , Ion Channel Gating , Models, Molecular , Molecular Dynamics Simulation , Mutation/genetics , Myocardium , NAV1.5 Voltage-Gated Sodium Channel/isolation & purification , NAV1.5 Voltage-Gated Sodium Channel/ultrastructure , Propafenone/pharmacology , Protein Conformation , Rats , Sodium/metabolism , Time Factors , Water/chemistry
7.
Cell ; 176(4): 913-927.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30686581

ABSTRACT

Tissue engineering using cardiomyocytes derived from human pluripotent stem cells holds a promise to revolutionize drug discovery, but only if limitations related to cardiac chamber specification and platform versatility can be overcome. We describe here a scalable tissue-cultivation platform that is cell source agnostic and enables drug testing under electrical pacing. The plastic platform enabled on-line noninvasive recording of passive tension, active force, contractile dynamics, and Ca2+ transients, as well as endpoint assessments of action potentials and conduction velocity. By combining directed cell differentiation with electrical field conditioning, we engineered electrophysiologically distinct atrial and ventricular tissues with chamber-specific drug responses and gene expression. We report, for the first time, engineering of heteropolar cardiac tissues containing distinct atrial and ventricular ends, and we demonstrate their spatially confined responses to serotonin and ranolazine. Uniquely, electrical conditioning for up to 8 months enabled modeling of polygenic left ventricular hypertrophy starting from patient cells.


Subject(s)
Myocytes, Cardiac/cytology , Tissue Culture Techniques/instrumentation , Tissue Engineering/methods , Action Potentials , Cell Differentiation , Cells, Cultured , Electrophysiological Phenomena , Humans , Induced Pluripotent Stem Cells/cytology , Models, Biological , Myocardium/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Tissue Culture Techniques/methods
8.
Immunity ; 56(10): 2342-2357.e10, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37625409

ABSTRACT

The heart is an autoimmune-prone organ. It is crucial for the heart to keep injury-induced autoimmunity in check to avoid autoimmune-mediated inflammatory disease. However, little is known about how injury-induced autoimmunity is constrained in hearts. Here, we reveal an unknown intramyocardial immunosuppressive program driven by Tbx1, a DiGeorge syndrome disease gene that encodes a T-box transcription factor (TF). We found induced profound lymphangiogenic and immunomodulatory gene expression changes in lymphatic endothelial cells (LECs) after myocardial infarction (MI). The activated LECs penetrated the infarcted area and functioned as intramyocardial immune hubs to increase the numbers of tolerogenic dendritic cells (tDCs) and regulatory T (Treg) cells through the chemokine Ccl21 and integrin Icam1, thereby inhibiting the expansion of autoreactive CD8+ T cells and promoting reparative macrophage expansion to facilitate post-MI repair. Mimicking its timing and implementation may be an additional approach to treating autoimmunity-mediated cardiac diseases.

9.
Cell ; 171(3): 573-587.e14, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033129

ABSTRACT

Progenitor cells differentiate into specialized cell types through coordinated expression of lineage-specific genes and modification of complex chromatin configurations. We demonstrate that a histone deacetylase (Hdac3) organizes heterochromatin at the nuclear lamina during cardiac progenitor lineage restriction. Specification of cardiomyocytes is associated with reorganization of peripheral heterochromatin, and independent of deacetylase activity, Hdac3 tethers peripheral heterochromatin containing lineage-relevant genes to the nuclear lamina. Deletion of Hdac3 in cardiac progenitor cells releases genomic regions from the nuclear periphery, leading to precocious cardiac gene expression and differentiation into cardiomyocytes; in contrast, restricting Hdac3 to the nuclear periphery rescues myogenesis in progenitors otherwise lacking Hdac3. Our results suggest that availability of genomic regions for activation by lineage-specific factors is regulated in part through dynamic chromatin-nuclear lamina interactions and that competence of a progenitor cell to respond to differentiation signals may depend upon coordinated movement of responding gene loci away from the nuclear periphery.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Nuclear Lamina/metabolism , Stem Cells/cytology , Animals , Genome , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cells/metabolism
10.
Cell ; 168(1-2): 111-120.e11, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28086084

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels underlie the control of rhythmic activity in cardiac and neuronal pacemaker cells. In HCN, the polarity of voltage dependence is uniquely reversed. Intracellular cyclic adenosine monophosphate (cAMP) levels tune the voltage response, enabling sympathetic nerve stimulation to increase the heart rate. We present cryo-electron microscopy structures of the human HCN channel in the absence and presence of cAMP at 3.5 Å resolution. HCN channels contain a K+ channel selectivity filter-forming sequence from which the amino acids create a unique structure that explains Na+ and K+ permeability. The voltage sensor adopts a depolarized conformation, and the pore is closed. An S4 helix of unprecedented length extends into the cytoplasm, contacts the C-linker, and twists the inner helical gate shut. cAMP binding rotates cytoplasmic domains to favor opening of the inner helical gate. These structures advance understanding of ion selectivity, reversed polarity gating, and cAMP regulation in HCN channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Potassium Channels/chemistry , Amino Acid Sequence , Cryoelectron Microscopy/methods , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Models, Molecular , Potassium Channels/metabolism , Sequence Alignment
11.
Physiol Rev ; 104(3): 1265-1333, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38153307

ABSTRACT

The complexity of cardiac electrophysiology, involving dynamic changes in numerous components across multiple spatial (from ion channel to organ) and temporal (from milliseconds to days) scales, makes an intuitive or empirical analysis of cardiac arrhythmogenesis challenging. Multiscale mechanistic computational models of cardiac electrophysiology provide precise control over individual parameters, and their reproducibility enables a thorough assessment of arrhythmia mechanisms. This review provides a comprehensive analysis of models of cardiac electrophysiology and arrhythmias, from the single cell to the organ level, and how they can be leveraged to better understand rhythm disorders in cardiac disease and to improve heart patient care. Key issues related to model development based on experimental data are discussed, and major families of human cardiomyocyte models and their applications are highlighted. An overview of organ-level computational modeling of cardiac electrophysiology and its clinical applications in personalized arrhythmia risk assessment and patient-specific therapy of atrial and ventricular arrhythmias is provided. The advancements presented here highlight how patient-specific computational models of the heart reconstructed from patient data have achieved success in predicting risk of sudden cardiac death and guiding optimal treatments of heart rhythm disorders. Finally, an outlook toward potential future advances, including the combination of mechanistic modeling and machine learning/artificial intelligence, is provided. As the field of cardiology is embarking on a journey toward precision medicine, personalized modeling of the heart is expected to become a key technology to guide pharmaceutical therapy, deployment of devices, and surgical interventions.


Subject(s)
Arrhythmias, Cardiac , Models, Cardiovascular , Humans , Arrhythmias, Cardiac/physiopathology , Animals , Computer Simulation , Translational Research, Biomedical , Myocytes, Cardiac/physiology , Electrophysiological Phenomena/physiology , Action Potentials/physiology
12.
Physiol Rev ; 104(3): 931-982, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38300522

ABSTRACT

Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.


Subject(s)
Cardiovascular Diseases , Proteomics , Animals , Humans , Proteomics/methods , Heart , Protein Processing, Post-Translational , Mass Spectrometry/methods
13.
Physiol Rev ; 103(3): 2271-2319, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36731030

ABSTRACT

The intercalated disc (ID) is a highly specialized structure that connects cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery and an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance of normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.


Subject(s)
Myocardium , Myocytes, Cardiac , Humans , Myocytes, Cardiac/physiology , Myocardium/metabolism , Gap Junctions/metabolism , Arrhythmias, Cardiac
14.
Physiol Rev ; 103(1): 391-432, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35953269

ABSTRACT

The heart is imbued with a vast lymphatic network that is responsible for fluid homeostasis and immune cell trafficking. Disturbances in the forces that regulate microvascular fluid movement can result in myocardial edema, which has profibrotic and proinflammatory consequences and contributes to cardiovascular dysfunction. This review explores the complex relationship between cardiac lymphatics, myocardial edema, and cardiac disease. It covers the revised paradigm of microvascular forces and fluid movement around the capillary as well as the arsenal of preclinical tools and animal models used to model myocardial edema and cardiac disease. Clinical studies of myocardial edema and their prognostic significance are examined in parallel to the recent elegant animal studies discerning the pathophysiological role and therapeutic potential of cardiac lymphatics in different cardiovascular disease models. This review highlights the outstanding questions of interest to both basic scientists and clinicians regarding the roles of cardiac lymphatics in health and disease.


Subject(s)
Edema, Cardiac , Heart Diseases , Lymphatic Vessels , Animals , Disease Models, Animal , Edema, Cardiac/physiopathology , Heart Diseases/physiopathology , Lymphatic Vessels/physiopathology
15.
Immunity ; 54(9): 2057-2071.e6, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34363749

ABSTRACT

Hypertension affects one-third of the world's population, leading to cardiac dysfunction that is modulated by resident and recruited immune cells. Cardiomyocyte growth and increased cardiac mass are essential to withstand hypertensive stress; however, whether immune cells are involved in this compensatory cardioprotective process is unclear. In normotensive animals, single-cell transcriptomics of fate-mapped self-renewing cardiac resident macrophages (RMs) revealed transcriptionally diverse cell states with a core repertoire of reparative gene programs, including high expression of insulin-like growth factor-1 (Igf1). Hypertension drove selective in situ proliferation and transcriptional activation of some cardiac RM states, directly correlating with increased cardiomyocyte growth. During hypertension, inducible ablation of RMs or selective deletion of RM-derived Igf1 prevented adaptive cardiomyocyte growth, and cardiac mass failed to increase, which led to cardiac dysfunction. Single-cell transcriptomics identified a conserved IGF1-expressing macrophage subpopulation in human cardiomyopathy. Here we defined the absolute requirement of RM-produced IGF-1 in cardiac adaptation to hypertension.


Subject(s)
Adaptation, Physiological/physiology , Hypertension/metabolism , Insulin-Like Growth Factor I/metabolism , Macrophages/metabolism , Ventricular Remodeling/physiology , Animals , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Hypertension/complications , Hypertension/immunology , Infant , Male , Mice , Middle Aged , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology
16.
Immunity ; 54(9): 2072-2088.e7, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34320366

ABSTRACT

Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2+) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2-) macrophages. Herein, we identified an essential role for CCR2- macrophages in the chronically failing heart. Depletion of CCR2- macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2- macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4)-dependent pathway that controlled growth factor expression. These findings establish a role for tissue-resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Ventricular Remodeling/physiology , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Myocardium/metabolism , Troponin T/genetics
17.
Mol Cell ; 82(19): 3661-3676.e8, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206740

ABSTRACT

Mitochondrial Ca2+ uptake, mediated by the mitochondrial Ca2+ uniporter, regulates oxidative phosphorylation, apoptosis, and intracellular Ca2+ signaling. Previous studies suggest that non-neuronal uniporters are exclusively regulated by a MICU1-MICU2 heterodimer. Here, we show that skeletal-muscle and kidney uniporters also complex with a MICU1-MICU1 homodimer and that human/mouse cardiac uniporters are largely devoid of MICUs. Cells employ protein-importation machineries to fine-tune the relative abundance of MICU1 homo- and heterodimers and utilize a conserved MICU intersubunit disulfide to protect properly assembled dimers from proteolysis by YME1L1. Using the MICU1 homodimer or removing MICU1 allows mitochondria to more readily take up Ca2+ so that cells can produce more ATP in response to intracellular Ca2+ transients. However, the trade-off is elevated ROS, impaired basal metabolism, and higher susceptibility to death. These results provide mechanistic insights into how tissues can manipulate mitochondrial Ca2+ uptake properties to support their unique physiological functions.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium , Cation Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Adenosine Triphosphate , Animals , Calcium/metabolism , Calcium Channels , Calcium-Binding Proteins/genetics , Disulfides/metabolism , Humans , Mice , Mitochondrial Membrane Transport Proteins/genetics , Reactive Oxygen Species/metabolism
18.
Genes Dev ; 36(7-8): 468-482, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35450884

ABSTRACT

The nucleosome remodeling and deacetylase (NuRD) complex is one of the central chromatin remodeling complexes that mediates gene repression. NuRD is essential for numerous developmental events, including heart development. Clinical and genetic studies have provided direct evidence for the role of chromodomain helicase DNA-binding protein 4 (CHD4), the catalytic component of NuRD, in congenital heart disease (CHD), including atrial and ventricular septal defects. Furthermore, it has been demonstrated that CHD4 is essential for mammalian cardiomyocyte formation and function. A key unresolved question is how CHD4/NuRD is localized to specific cardiac target genes, as neither CHD4 nor NuRD can directly bind DNA. Here, we coupled a bioinformatics-based approach with mass spectrometry analyses to demonstrate that CHD4 interacts with the core cardiac transcription factors GATA4, NKX2-5, and TBX5 during embryonic heart development. Using transcriptomics and genome-wide occupancy data, we characterized the genomic landscape of GATA4, NKX2-5, and TBX5 repression and defined the direct cardiac gene targets of the GATA4-CHD4, NKX2-5-CHD4, and TBX5-CHD4 complexes. These data were used to identify putative cis-regulatory elements controlled by these complexes. We genetically interrogated two of these silencers in vivo: Acta1 and Myh11 We show that deletion of these silencers leads to inappropriate skeletal and smooth muscle gene misexpression, respectively, in the embryonic heart. These results delineate how CHD4/NuRD is localized to specific cardiac loci and explicates how mutations in the broadly expressed CHD4 protein lead to cardiac-specific disease states.


Subject(s)
DNA Helicases , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Animals , DNA Helicases/metabolism , Genes, Homeobox , Mammals/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Myocytes, Cardiac/metabolism , Nucleosomes , Transcription Factors/genetics
19.
Physiol Rev ; 101(1): 37-92, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32380895

ABSTRACT

The heart is vital for biological function in almost all chordates, including humans. It beats continually throughout our life, supplying the body with oxygen and nutrients while removing waste products. If it stops, so does life. The heartbeat involves precise coordination of the activity of billions of individual cells, as well as their swift and well-coordinated adaption to changes in physiological demand. Much of the vital control of cardiac function occurs at the level of individual cardiac muscle cells, including acute beat-by-beat feedback from the local mechanical environment to electrical activity (as opposed to longer term changes in gene expression and functional or structural remodeling). This process is known as mechano-electric coupling (MEC). In the current review, we present evidence for, and implications of, MEC in health and disease in human; summarize our understanding of MEC effects gained from whole animal, organ, tissue, and cell studies; identify potential molecular mediators of MEC responses; and demonstrate the power of computational modeling in developing a more comprehensive understanding of ?what makes the heart tick.Ë®.


Subject(s)
Heart Rate/physiology , Heart/physiology , Physical Stimulation , Animals , Arrhythmias, Cardiac/physiopathology , Biological Clocks , Humans , Myocardium/cytology , Myocytes, Cardiac/physiology
20.
Mol Cell ; 80(1): 59-71.e4, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32818430

ABSTRACT

Cardiac disease remains the leading cause of morbidity and mortality worldwide. The ß1-adrenergic receptor (ß1-AR) is a major regulator of cardiac functions and is downregulated in the majority of heart failure cases. A key physiological process is the activation of heterotrimeric G-protein Gs by ß1-ARs, leading to increased heart rate and contractility. Here, we use cryo-electron microscopy and functional studies to investigate the molecular mechanism by which ß1-AR activates Gs. We find that the tilting of α5-helix breaks a hydrogen bond between the sidechain of His373 in the C-terminal α5-helix and the backbone carbonyl of Arg38 in the N-terminal αN-helix of Gαs. Together with the disruption of another interacting network involving Gln59 in the α1-helix, Ala352 in the ß6-α5 loop, and Thr355 in the α5-helix, these conformational changes might lead to the deformation of the GDP-binding pocket. Our data provide molecular insights into the activation of G-proteins by G-protein-coupled receptors.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Isoproterenol/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Animals , Binding Sites , Cattle , Cell Line , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL