Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67.613
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 184(8): 2167-2182.e22, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33811809

ABSTRACT

Cardiac injury and dysfunction occur in COVID-19 patients and increase the risk of mortality. Causes are ill defined but could be through direct cardiac infection and/or inflammation-induced dysfunction. To identify mechanisms and cardio-protective drugs, we use a state-of-the-art pipeline combining human cardiac organoids with phosphoproteomics and single nuclei RNA sequencing. We identify an inflammatory "cytokine-storm", a cocktail of interferon gamma, interleukin 1ß, and poly(I:C), induced diastolic dysfunction. Bromodomain-containing protein 4 is activated along with a viral response that is consistent in both human cardiac organoids (hCOs) and hearts of SARS-CoV-2-infected K18-hACE2 mice. Bromodomain and extraterminal family inhibitors (BETi) recover dysfunction in hCOs and completely prevent cardiac dysfunction and death in a mouse cytokine-storm model. Additionally, BETi decreases transcription of genes in the viral response, decreases ACE2 expression, and reduces SARS-CoV-2 infection of cardiomyocytes. Together, BETi, including the Food and Drug Administration (FDA) breakthrough designated drug, apabetalone, are promising candidates to prevent COVID-19 mediated cardiac damage.


Subject(s)
COVID-19/complications , Cardiotonic Agents/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Heart Diseases/drug therapy , Quinazolinones/therapeutic use , Transcription Factors/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Cytokines/metabolism , Female , Heart Diseases/etiology , Human Embryonic Stem Cells , Humans , Inflammation/complications , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Transcription Factors/metabolism , COVID-19 Drug Treatment
3.
Cell ; 173(6): 1323-1327, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856952

ABSTRACT

Tubulin posttranslational modifications are currently emerging as important regulators of the microtubule cytoskeleton and thus have a strong potential to be implicated in a number of disorders. Here, we review the latest advances in understanding the physiological roles of tubulin modifications and their links to a variety of pathologies.


Subject(s)
Protein Processing, Post-Translational , Tubulin/chemistry , Animals , Blood Platelets/metabolism , Cilia/metabolism , Cytoskeleton/metabolism , Flagella/metabolism , Heart Diseases/metabolism , Humans , Mice , Microtubules/metabolism , Mutation , Neurodegenerative Diseases/therapy , Phenotype , Risk Factors , Tubulin/physiology
4.
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
5.
Nature ; 627(8005): 854-864, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480880

ABSTRACT

The heart, which is the first organ to develop, is highly dependent on its form to function1,2. However, how diverse cardiac cell types spatially coordinate to create the complex morphological structures that are crucial for heart function remains unclear. Here we integrated single-cell RNA-sequencing with high-resolution multiplexed error-robust fluorescence in situ hybridization to resolve the identity of the cardiac cell types that develop the human heart. This approach also provided a spatial mapping of individual cells that enables illumination of their organization into cellular communities that form distinct cardiac structures. We discovered that many of these cardiac cell types further specified into subpopulations exclusive to specific communities, which support their specialization according to the cellular ecosystem and anatomical region. In particular, ventricular cardiomyocyte subpopulations displayed an unexpected complex laminar organization across the ventricular wall and formed, with other cell subpopulations, several cellular communities. Interrogating cell-cell interactions within these communities using in vivo conditional genetic mouse models and in vitro human pluripotent stem cell systems revealed multicellular signalling pathways that orchestrate the spatial organization of cardiac cell subpopulations during ventricular wall morphogenesis. These detailed findings into the cellular social interactions and specialization of cardiac cell types constructing and remodelling the human heart offer new insights into structural heart diseases and the engineering of complex multicellular tissues for human heart repair.


Subject(s)
Body Patterning , Heart , Myocardium , Animals , Humans , Mice , Heart/anatomy & histology , Heart/embryology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/embryology , In Situ Hybridization, Fluorescence , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Gene Expression Analysis
6.
Nature ; 619(7971): 707-715, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37495877

ABSTRACT

Engineering a patient's own T cells to selectively target and eliminate tumour cells has cured patients with untreatable haematologic cancers. These results have energized the field to apply chimaeric antigen receptor (CAR) T therapy throughout oncology. However, evidence from clinical and preclinical studies underscores the potential of CAR T therapy beyond oncology in treating autoimmunity, chronic infections, cardiac fibrosis, senescence-associated disease and other conditions. Concurrently, the deployment of new technologies and platforms provides further opportunity for the application of CAR T therapy to noncancerous pathologies. Here we review the rationale behind CAR T therapy, current challenges faced in oncology, a synopsis of preliminary reports in noncancerous diseases, and a discussion of relevant emerging technologies. We examine potential applications for this therapy in a wide range of contexts. Last, we highlight concerns regarding specificity and safety and outline the path forward for CAR T therapy beyond cancer.


Subject(s)
Aging , Autoimmune Diseases , Fibrosis , Heart Diseases , Immunotherapy, Adoptive , Infections , Neoplasms , Receptors, Chimeric Antigen , Humans , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Neoplasms/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes/immunology , Autoimmune Diseases/therapy , Infections/therapy , Fibrosis/therapy , Aging/pathology , Heart Diseases/therapy
7.
Annu Rev Cell Dev Biol ; 30: 535-60, 2014.
Article in English | MEDLINE | ID: mdl-25062362

ABSTRACT

Although most modern dog breeds are less than 200 years old, the symbiosis between man and dog is ancient. Since prehistoric times, repeated selection events have transformed the wolf into man's guardians, laborers, athletes, and companions. The rapid transformation from pack predator to loyal companion is a feat that is arguably unique among domesticated animals. How this transformation came to pass remained a biological mystery until recently: Within the past decade, the deployment of genomic approaches to study population structure, detect signatures of selection, and identify genetic variants that underlie canine phenotypes is ushering into focus novel biological mechanisms that make dogs remarkable. Ironically, the very practices responsible for breed formation also spurned morbidity; today, many diseases are correlated with breed identity. In this review, we discuss man's best friend in the context of a genetic model to understand paradigms of heritable phenotypes, both desirable and disadvantageous.


Subject(s)
Dogs/genetics , Genome , Animals , Body Size/genetics , Bone Neoplasms/genetics , Bone Neoplasms/veterinary , Breeding , Chromosome Mapping , Disease Models, Animal , Dog Diseases/genetics , Dogs/anatomy & histology , Dogs/classification , Extremities/anatomy & histology , Genome-Wide Association Study , Glycoproteins/genetics , Glycoproteins/physiology , HMGA2 Protein/genetics , HMGA2 Protein/physiology , Hair/anatomy & histology , Heart Diseases/genetics , Heart Diseases/veterinary , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/veterinary , Osteosarcoma/genetics , Osteosarcoma/veterinary , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Selection, Genetic , Skin/anatomy & histology , Skull/anatomy & histology , Smad2 Protein/genetics , Smad2 Protein/physiology , Species Specificity , Tail/anatomy & histology
8.
Physiol Rev ; 100(2): 673-694, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31751167

ABSTRACT

The central dogma of molecular biology illustrates the importance of mRNAs as critical mediators between genetic information encoded at the DNA level and proteomes/metabolomes that determine the diverse functional outcome at the cellular and organ levels. Although the total number of protein-producing (coding) genes in the mammalian genome is ~20,000, it is evident that the intricate processes of cardiac development and the highly regulated physiological regulation in the normal heart, as well as the complex manifestation of pathological remodeling in a diseased heart, would require a much higher degree of complexity at the transcriptome level and beyond. Indeed, in addition to an extensive regulatory scheme implemented at the level of transcription, the complexity of transcript processing following transcription is dramatically increased. RNA processing includes post-transcriptional modification, alternative splicing, editing and transportation, ribosomal loading, and degradation. While transcriptional control of cardiac genes has been a major focus of investigation in recent decades, a great deal of progress has recently been made in our understanding of how post-transcriptional regulation of mRNA contributes to transcriptome complexity. In this review, we highlight some of the key molecular processes and major players in RNA maturation and post-transcriptional regulation. In addition, we provide an update to the recent progress made in the discovery of RNA processing regulators implicated in cardiac development and disease. While post-transcriptional modulation is a complex and challenging problem to study, recent technological advancements are paving the way for a new era of exciting discoveries and potential clinical translation in the context of cardiac biology and heart disease.


Subject(s)
Heart Diseases/metabolism , Heart/embryology , Myocardium/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Animals , Gene Expression Regulation, Developmental , Heart Diseases/drug therapy , Heart Diseases/genetics , Heart Diseases/pathology , Humans , Morphogenesis , Myocardium/pathology , RNA Stability , RNA, Messenger/genetics , Transcription, Genetic
9.
Nature ; 595(7867): 438-443, 2021 07.
Article in English | MEDLINE | ID: mdl-34163071

ABSTRACT

In diseased organs, stress-activated signalling cascades alter chromatin, thereby triggering maladaptive cell state transitions. Fibroblast activation is a common stress response in tissues that worsens lung, liver, kidney and heart disease, yet its mechanistic basis remains unclear1,2. Pharmacological inhibition of bromodomain and extra-terminal domain (BET) proteins alleviates cardiac dysfunction3-7, providing a tool to interrogate and modulate cardiac cell states as a potential therapeutic approach. Here we use single-cell epigenomic analyses of hearts dynamically exposed to BET inhibitors to reveal a reversible transcriptional switch that underlies the activation of fibroblasts. Resident cardiac fibroblasts demonstrated robust toggling between the quiescent and activated state in a manner directly correlating with BET inhibitor exposure and cardiac function. Single-cell chromatin accessibility revealed previously undescribed DNA elements, the accessibility of which dynamically correlated with cardiac performance. Among the most dynamic elements was an enhancer that regulated the transcription factor MEOX1, which was specifically expressed in activated fibroblasts, occupied putative regulatory elements of a broad fibrotic gene program and was required for TGFß-induced fibroblast activation. Selective CRISPR inhibition of the single most dynamic cis-element within the enhancer blocked TGFß-induced Meox1 activation. We identify MEOX1 as a central regulator of fibroblast activation associated with cardiac dysfunction and demonstrate its upregulation after activation of human lung, liver and kidney fibroblasts. The plasticity and specificity of BET-dependent regulation of MEOX1 in tissue fibroblasts provide previously unknown trans- and cis-targets for treating fibrotic disease.


Subject(s)
Enhancer Elements, Genetic , Fibroblasts/cytology , Heart Diseases/genetics , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Epigenomics , Gene Expression Regulation , Humans , Mice , Proteins/antagonists & inhibitors , Single-Cell Analysis , Transcriptome , Transforming Growth Factor beta/metabolism
10.
Semin Cell Dev Biol ; 156: 190-200, 2024 03 15.
Article in English | MEDLINE | ID: mdl-36641366

ABSTRACT

The parasympathetic nervous system via the vagus nerve exerts profound influence over the heart. Together with the sympathetic nervous system, the parasympathetic nervous system is responsible for fine-tuned regulation of all aspects of cardiovascular function, including heart rate, rhythm, contractility, and blood pressure. In this review, we highlight vagal efferent and afferent innervation of the heart, with a focus on insights from comparative biology and advances in understanding the molecular and genetic diversity of vagal neurons, as well as interoception, parasympathetic dysfunction in heart disease, and the therapeutic potential of targeting the parasympathetic nervous system in cardiovascular disease.


Subject(s)
Clinical Medicine , Heart Diseases , Humans , Vagus Nerve/physiology , Heart , Heart Rate/physiology
11.
Physiol Rev ; 99(1): 381-426, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30379622

ABSTRACT

The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.


Subject(s)
Carrier Proteins/metabolism , Heart Diseases/metabolism , Heart Diseases/physiopathology , Myocardium/metabolism , Sarcomeres/metabolism , Animals , Heart Diseases/genetics , Humans , Mutation/genetics , Phosphorylation/physiology
12.
Physiol Rev ; 99(4): 1765-1817, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31364924

ABSTRACT

Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.


Subject(s)
Cell Death , Cytotoxicity, Immunologic , Heart Diseases/pathology , Mitochondria, Heart/pathology , Myocardium/pathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Autophagy , Autophagy-Related Proteins/metabolism , Heart Diseases/immunology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Mitochondria, Heart/immunology , Mitochondria, Heart/metabolism , Myocardium/immunology , Myocardium/metabolism , Necrosis , Pyroptosis , Signal Transduction
13.
N Engl J Med ; 388(4): 319-332, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36511784

ABSTRACT

BACKGROUND: In a multinational, phase 3, head-to-head trial, ibrutinib, a Bruton's tyrosine kinase (BTK) inhibitor, was compared with zanubrutinib, a BTK inhibitor with greater specificity, as treatment for relapsed or refractory chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL). In prespecified interim analyses, zanubrutinib was superior to ibrutinib with respect to overall response (the primary end point). Data from the final analysis of progression-free survival are now available. METHODS: We randomly assigned, in a 1:1 ratio, patients with relapsed or refractory CLL or SLL who had received at least one previous course of therapy to receive zanubrutinib or ibrutinib until the occurrence of disease progression or unacceptable toxic effects. In this final analysis, progression-free survival (a key secondary end point) was assessed with the use of a hierarchical testing strategy to determine whether zanubrutinib was noninferior to ibrutinib. If noninferiority was established, the superiority of zanubrutinib was assessed and claimed if the two-sided P value was less than 0.05. RESULTS: At a median follow-up of 29.6 months, zanubrutinib was found to be superior to ibrutinib with respect to progression-free survival among 652 patients (hazard ratio for disease progression or death, 0.65; 95% confidence interval, [CI], 0.49 to 0.86; P = 0.002), as assessed by the investigators; the results were similar to those as assessed by an independent-review committee. At 24 months, the investigator-assessed rates of progression-free survival were 78.4% in the zanubrutinib group and 65.9% in the ibrutinib group. Among patients with a 17p deletion, a TP53 mutation, or both, those who received zanubrutinib had longer progression-free survival than those who received ibrutinib (hazard ratio for disease progression or death, 0.53; 95% CI, 0.31 to 0.88); progression-free survival across other major subgroups consistently favored zanubrutinib. The percentage of patients with an overall response was higher in the zanubrutinib group than in the ibrutinib group. The safety profile of zanubrutinib was better than that of ibrutinib, with fewer adverse events leading to treatment discontinuation and fewer cardiac events, including fewer cardiac events leading to treatment discontinuation or death. CONCLUSIONS: In patients with relapsed or refractory CLL or SLL, progression-free survival was significantly longer among patients who received zanubrutinib than among those who received ibrutinib, and zanubrutinib was associated with fewer cardiac adverse events. (Funded by BeiGene; ALPINE ClinicalTrials.gov number, NCT03734016.).


Subject(s)
Antineoplastic Agents , Heart Diseases , Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Disease Progression , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Heart Diseases/chemically induced
14.
Circ Res ; 134(12): 1663-1680, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843286

ABSTRACT

Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.


Subject(s)
Heart Diseases , Humans , Animals , Heart Diseases/immunology , Heart Diseases/therapy , Allergy and Immunology/trends , Allergy and Immunology/history , Cardiovascular Diseases/immunology , History, 21st Century , History, 20th Century
15.
Circ Res ; 134(11): 1636-1660, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781295

ABSTRACT

Contemporary World Health Organization data indicates that ≈39 million people are living with the human immunodeficiency virus. Of these, 24 million have been reported to have successfully accessed combination antiretroviral therapy. In 1996, the World Health Organization endorsed the widespread use of combination antiretroviral therapy, transforming human immunodeficiency virus infection from being a life-threatening disease to a chronic illness characterized by multiple comorbidities. The increased access to combination antiretroviral therapy has translated to people living with human immunodeficiency virus (PLWH) no longer having a reduced life expectancy. Although aging as a biological process increases exposure to oxidative stress and subsequent systemic inflammation, this effect is likely enhanced in PLWH as they age. This narrative review engages the intricate interplay between human immunodeficiency virus associated chronic inflammation, combination antiretroviral therapy, and cardiac and renal comorbidities development in aging PLWH. We examine the evolving demographic profile of PLWH, emphasizing the increasing prevalence of aging individuals within this population. A central focus of the review discusses the pathophysiological mechanisms that underpin the heightened susceptibility of PLWH to renal and cardiac diseases as they age.


Subject(s)
Aging , Comorbidity , HIV Infections , Humans , HIV Infections/epidemiology , HIV Infections/drug therapy , Kidney Diseases/epidemiology , Heart Diseases/epidemiology , Aged
16.
Circ Res ; 134(4): 459-473, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38359096

ABSTRACT

Hypertension is the leading cause of cardiovascular disease in women, and sub-Saharan African (SSA) countries have some of the highest rates of hypertension in the world. Expanding knowledge of causes, management, and awareness of hypertension and its co-morbidities worldwide is an effective strategy to mitigate its harms, decrease morbidities and mortality, and improve individual quality of life. Hypertensive disorders of pregnancy (HDPs) are a particularly important subset of hypertension, as pregnancy is a major stress test of the cardiovascular system and can be the first instance in which cardiovascular disease is clinically apparent. In SSA, women experience a higher incidence of HDP compared with other African regions. However, the region has yet to adopt treatment and preventative strategies for HDP. This delay stems from insufficient awareness, lack of clinical screening for hypertension, and lack of prevention programs. In this brief literature review, we will address the long-term consequences of hypertension and HDP in women. We evaluate the effects of uncontrolled hypertension in SSA by including research on heart disease, stroke, kidney disease, peripheral arterial disease, and HDP. Limitations exist in the number of studies from SSA; therefore, we will use data from countries across the globe, comparing and contrasting approaches in similar and dissimilar populations. Our review highlights an urgent need to prioritize public health, clinical, and bench research to discover cost-effective preventative and treatment strategies that will improve the lives of women living with hypertension in SSA.


Subject(s)
Cardiovascular Diseases , Heart Diseases , Hypertension, Pregnancy-Induced , Hypertension , Pregnancy , Humans , Female , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/therapy , Quality of Life , Hypertension/diagnosis , Hypertension/epidemiology , Africa South of the Sahara/epidemiology
17.
Circ Res ; 134(8): 1006-1022, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38506047

ABSTRACT

BACKGROUND: In heart failure, signaling downstream the ß2-adrenergic receptor is critical. Sympathetic stimulation of ß2-adrenergic receptor alters cAMP (cyclic adenosine 3',5'-monophosphate) and triggers PKA (protein kinase A)-dependent phosphorylation of proteins that regulate cardiac function. cAMP levels are regulated in part by PDEs (phosphodiesterases). Several AKAPs (A kinase anchoring proteins) regulate cardiac function and are proposed as targets for precise pharmacology. AKAP12 is expressed in the heart and has been reported to directly bind ß2-adrenergic receptor, PKA, and PDE4D. However, its roles in cardiac function are unclear. METHODS: cAMP accumulation in real time downstream of the ß2-adrenergic receptor was detected for 60 minutes in live cells using the luciferase-based biosensor (GloSensor) in AC16 human-derived cardiomyocyte cell lines overexpressing AKAP12 versus controls. Cardiomyocyte intracellular calcium and contractility were studied in adult primary cardiomyocytes from male and female mice overexpressing cardiac AKAP12 (AKAP12OX) and wild-type littermates post acute treatment with 100-nM isoproterenol (ISO). Systolic cardiac function was assessed in mice after 14 days of subcutaneous ISO administration (60 mg/kg per day). AKAP12 gene and protein expression levels were evaluated in left ventricular samples from patients with end-stage heart failure. RESULTS: AKAP12 upregulation significantly reduced total intracellular cAMP levels in AC16 cells through PDE8. Adult primary cardiomyocytes from AKAP12OX mice had significantly reduced contractility and impaired calcium handling in response to ISO, which was reversed in the presence of the selective PDE8 inhibitor (PF-04957325). AKAP12OX mice had deteriorated systolic cardiac function and enlarged left ventricles. Patients with end-stage heart failure had upregulated gene and protein levels of AKAP12. CONCLUSIONS: AKAP12 upregulation in cardiac tissue is associated with accelerated cardiac dysfunction through the AKAP12-PDE8 axis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Heart Diseases , Receptors, Adrenergic , Animals , Female , Humans , Male , Mice , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Calcium/metabolism , Cell Cycle Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Diseases/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Isoproterenol/pharmacology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic/metabolism , Up-Regulation
18.
Nature ; 580(7802): 252-256, 2020 04.
Article in English | MEDLINE | ID: mdl-32269341

ABSTRACT

Accurate assessment of cardiac function is crucial for the diagnosis of cardiovascular disease1, screening for cardiotoxicity2 and decisions regarding the clinical management of patients with a critical illness3. However, human assessment of cardiac function focuses on a limited sampling of cardiac cycles and has considerable inter-observer variability despite years of training4,5. Here, to overcome this challenge, we present a video-based deep learning algorithm-EchoNet-Dynamic-that surpasses the performance of human experts in the critical tasks of segmenting the left ventricle, estimating ejection fraction and assessing cardiomyopathy. Trained on echocardiogram videos, our model accurately segments the left ventricle with a Dice similarity coefficient of 0.92, predicts ejection fraction with a mean absolute error of 4.1% and reliably classifies heart failure with reduced ejection fraction (area under the curve of 0.97). In an external dataset from another healthcare system, EchoNet-Dynamic predicts the ejection fraction with a mean absolute error of 6.0% and classifies heart failure with reduced ejection fraction with an area under the curve of 0.96. Prospective evaluation with repeated human measurements confirms that the model has variance that is comparable to or less than that of human experts. By leveraging information across multiple cardiac cycles, our model can rapidly identify subtle changes in ejection fraction, is more reproducible than human evaluation and lays the foundation for precise diagnosis of cardiovascular disease in real time. As a resource to promote further innovation, we also make publicly available a large dataset of 10,030 annotated echocardiogram videos.


Subject(s)
Deep Learning , Heart Diseases/diagnosis , Heart Diseases/physiopathology , Heart/physiology , Heart/physiopathology , Models, Cardiovascular , Video Recording , Atrial Fibrillation , Datasets as Topic , Echocardiography , Heart Failure/physiopathology , Hospitals , Humans , Prospective Studies , Reproducibility of Results , Ventricular Function, Left/physiology
19.
Semin Immunol ; 59: 101602, 2022 01.
Article in English | MEDLINE | ID: mdl-35277300

ABSTRACT

Formyl peptide receptor type 2 (FPR2) regulates the initiation and resolution phases of the inflammatory response. In the setting of heart injury and disease, dysregulated inflammation can potentiate maladaptive healing and pathological remodeling of the heart leading to cardiac dysfunction and failure. The potential to regulate and resolve adverse inflammation is postulated to improve outcome in the setting of heart disease. This review covers emerging concepts on the role of FPR2 in heart disease and strategies to activate pro-resolution processes to limit disease progression. We summarize key preclinical studies that support use of FPR2 agonists in heart disease. Finally, we briefly discuss the status of FPR2 agonists under evaluation in the clinic.


Subject(s)
Heart Diseases , Receptors, Formyl Peptide , Humans , Inflammation/pathology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/physiology , Wound Healing
20.
Proc Natl Acad Sci U S A ; 120(37): e2309714120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669377

ABSTRACT

Proofreading (editing) of mischarged tRNAs by cytoplasmic aminoacyl-tRNA synthetases (aaRSs), whose impairment causes neurodegeneration and cardiac diseases, is of high significance for protein homeostasis. However, whether mitochondrial translation needs fidelity and the significance of editing by mitochondrial aaRSs have been unclear. Here, we show that mammalian cells critically depended on the editing of mitochondrial threonyl-tRNA synthetase (mtThrRS, encoded by Tars2), disruption of which accumulated Ser-tRNAThr and generated a large abundance of Thr-to-Ser misincorporated peptides in vivo. Such infidelity impaired mitochondrial translation and oxidative phosphorylation, causing oxidative stress and cell cycle arrest in the G0/G1 phase. Notably, reactive oxygen species (ROS) scavenging by N-acetylcysteine attenuated this abnormal cell proliferation. A mouse model of heart-specific defective mtThrRS editing was established. Increased ROS levels, blocked cardiomyocyte proliferation, contractile dysfunction, dilated cardiomyopathy, and cardiac fibrosis were observed. Our results elucidate that mitochondria critically require a high level of translational accuracy at Thr codons and highlight the cellular dysfunctions and imbalance in tissue homeostasis caused by mitochondrial mistranslation.


Subject(s)
Amino Acyl-tRNA Synthetases , Cardiomyopathies , Heart Diseases , Animals , Mice , Reactive Oxygen Species , Cell Cycle Checkpoints , Oxidative Stress , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL