Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.301
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30686582

RESUMEN

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Asunto(s)
Circulación Colateral/fisiología , Corazón/crecimiento & desarrollo , Regeneración/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Quimiocina CXCL12/metabolismo , Vasos Coronarios/crecimiento & desarrollo , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/fisiología , Receptores CXCR4/metabolismo , Transducción de Señal
2.
Cell ; 167(7): 1734-1749.e22, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984724

RESUMEN

Mutation of highly conserved residues in transcription factors may affect protein-protein or protein-DNA interactions, leading to gene network dysregulation and human disease. Human mutations in GATA4, a cardiogenic transcription factor, cause cardiac septal defects and cardiomyopathy. Here, iPS-derived cardiomyocytes from subjects with a heterozygous GATA4-G296S missense mutation showed impaired contractility, calcium handling, and metabolic activity. In human cardiomyocytes, GATA4 broadly co-occupied cardiac enhancers with TBX5, another transcription factor that causes septal defects when mutated. The GATA4-G296S mutation disrupted TBX5 recruitment, particularly to cardiac super-enhancers, concomitant with dysregulation of genes related to the phenotypic abnormalities, including cardiac septation. Conversely, the GATA4-G296S mutation led to failure of GATA4 and TBX5-mediated repression at non-cardiac genes and enhanced open chromatin states at endothelial/endocardial promoters. These results reveal how disease-causing missense mutations can disrupt transcriptional cooperativity, leading to aberrant chromatin states and cellular dysfunction, including those related to morphogenetic defects.


Asunto(s)
Factor de Transcripción GATA4/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Cromatina , Elementos de Facilitación Genéticos , Femenino , Corazón/crecimiento & desarrollo , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Mutación Missense , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética
3.
Cell ; 166(2): 451-467, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27419872

RESUMEN

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Asunto(s)
Mesodermo/citología , Transducción de Señal , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/citología , Huesos/metabolismo , Corazón/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/metabolismo , Análisis de la Célula Individual , Somitos/metabolismo , Células Madre , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo
4.
Cell ; 163(4): 1026-36, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26544945

RESUMEN

The magnitude of cardiomyocyte generation in the adult heart has been heavily debated. A recent report suggests that during mouse preadolescence, cardiomyocyte proliferation leads to a 40% increase in the number of cardiomyocytes. Such an expansion would change our understanding of heart growth and have far-reaching implications for cardiac regeneration. Here, using design-based stereology, we found that cardiomyocyte proliferation accounted for 30% of postnatal DNA synthesis; however, we were unable to detect any changes in cardiomyocyte number after postnatal day 11. (15)N-thymidine and BrdU analyses provided no evidence for a proliferative peak in preadolescent mice. By contrast, cardiomyocyte multinucleation comprises 57% of postnatal DNA synthesis, followed by cardiomyocyte nuclear polyploidisation, contributing with 13% to DNA synthesis within the second and third postnatal weeks. We conclude that the majority of cardiomyocytes is set within the first postnatal week and that this event is followed by two waves of non-replicative DNA synthesis. This Matters Arising paper is in response to Naqvi et al. (2014), published in Cell. See also the associated Correspondence by Soonpaa et al. (2015), and the response by Naqvi et al. (2015), published in this issue.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Animales , Masculino
5.
Cell ; 157(4): 765-7, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24813600

RESUMEN

Cardiomyocytes, the cells of the heart muscle, lose nearly all of their proliferative capacity after birth, limiting the heart's ability to regenerate. Naqvi et al. now identify a transient burst of cardiomyocyte proliferation during preadolescence, driven by a thyroid hormone surge, with therapeutic implications for congenital and acquired heart diseases.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Animales , Masculino
6.
Cell ; 157(4): 795-807, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24813607

RESUMEN

It is widely believed that perinatal cardiomyocyte terminal differentiation blocks cytokinesis, thereby causing binucleation and limiting regenerative repair after injury. This suggests that heart growth should occur entirely by cardiomyocyte hypertrophy during preadolescence when, in mice, cardiac mass increases many-fold over a few weeks. Here, we show that a thyroid hormone surge activates the IGF-1/IGF-1-R/Akt pathway on postnatal day 15 and initiates a brief but intense proliferative burst of predominantly binuclear cardiomyocytes. This proliferation increases cardiomyocyte numbers by ~40%, causing a major disparity between heart and cardiomyocyte growth. Also, the response to cardiac injury at postnatal day 15 is intermediate between that observed at postnatal days 2 and 21, further suggesting persistence of cardiomyocyte proliferative capacity beyond the perinatal period. If replicated in humans, this may allow novel regenerative therapies for heart diseases.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Animales , Separación Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/fisiología , Triyodotironina/metabolismo
7.
Nature ; 618(7964): 365-373, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225978

RESUMEN

Birth presents a metabolic challenge to cardiomyocytes as they reshape fuel preference from glucose to fatty acids for postnatal energy production1,2. This adaptation is triggered in part by post-partum environmental changes3, but the molecules orchestrating cardiomyocyte maturation remain unknown. Here we show that this transition is coordinated by maternally supplied γ-linolenic acid (GLA), an 18:3 omega-6 fatty acid enriched in the maternal milk. GLA binds and activates retinoid X receptors4 (RXRs), ligand-regulated transcription factors that are expressed in cardiomyocytes from embryonic stages. Multifaceted genome-wide analysis revealed that the lack of RXR in embryonic cardiomyocytes caused an aberrant chromatin landscape that prevented the induction of an RXR-dependent gene expression signature controlling mitochondrial fatty acid homeostasis. The ensuing defective metabolic transition featured blunted mitochondrial lipid-derived energy production and enhanced glucose consumption, leading to perinatal cardiac dysfunction and death. Finally, GLA supplementation induced RXR-dependent expression of the mitochondrial fatty acid homeostasis signature in cardiomyocytes, both in vitro and in vivo. Thus, our study identifies the GLA-RXR axis as a key transcriptional regulatory mechanism underlying the maternal control of perinatal cardiac metabolism.


Asunto(s)
Ácidos Grasos , Glucosa , Corazón , Leche Humana , Ácido gammalinolénico , Femenino , Humanos , Recién Nacido , Embarazo , Cromatina/genética , Ácidos Grasos/metabolismo , Ácido gammalinolénico/metabolismo , Ácido gammalinolénico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Corazón/efectos de los fármacos , Corazón/embriología , Corazón/crecimiento & desarrollo , Homeostasis , Técnicas In Vitro , Leche Humana/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Receptores X Retinoide/metabolismo , Factores de Transcripción/metabolismo
8.
Nature ; 599(7885): 431-435, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34789899

RESUMEN

A central question in chordate evolution is the origin of sessility in adult ascidians, and whether the appendicularian complete free-living style represents a primitive or derived condition among tunicates1. According to the 'a new heart for a new head' hypothesis, the evolution of the cardiopharyngeal gene regulatory network appears as a pivotal aspect to understand the evolution of the lifestyles of chordates2-4. Here we show that appendicularians experienced massive ancestral losses of cardiopharyngeal genes and subfunctions, leading to the 'deconstruction' of two ancestral modules of the tunicate cardiopharyngeal gene regulatory network. In ascidians, these modules are related to early and late multipotency, which is involved in lineage cell-fate determination towards the first and second heart fields and siphon muscles. Our work shows that the deconstruction of the cardiopharyngeal gene regulatory network involved the regressive loss of the siphon muscle, supporting an evolutionary scenario in which ancestral tunicates had a sessile ascidian-like adult lifestyle. In agreement with this scenario, our findings also suggest that this deconstruction contributed to the acceleration of cardiogenesis and the redesign of the heart into an open-wide laminar structure in appendicularians as evolutionary adaptations during their transition to a complete pelagic free-living style upon the innovation of the food-filtering house5.


Asunto(s)
Evolución Biológica , Corazón/anatomía & histología , Corazón/crecimiento & desarrollo , Urocordados/anatomía & histología , Urocordados/fisiología , Animales , Linaje de la Célula , Redes Reguladoras de Genes , Locomoción , Miocardio/citología , Miocardio/metabolismo , Urocordados/citología , Urocordados/genética
9.
Nature ; 585(7826): 563-568, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32939088

RESUMEN

Neural crest cells (NCCs) are migratory, multipotent embryonic cells that are unique to vertebrates and form an array of clade-defining adult features. The evolution of NCCs has been linked to various genomic events, including the evolution of new gene-regulatory networks1,2, the de novo evolution of genes3 and the proliferation of paralogous genes during genome-wide duplication events4. However, conclusive functional evidence linking new and/or duplicated genes to NCC evolution is lacking. Endothelin ligands (Edns) and endothelin receptors (Ednrs) are unique to vertebrates3,5,6, and regulate multiple aspects of NCC development in jawed vertebrates7-10. Here, to test whether the evolution of Edn signalling was a driver of NCC evolution, we used CRISPR-Cas9 mutagenesis11 to disrupt edn, ednr and dlx genes in the sea lamprey, Petromyzon marinus. Lampreys are jawless fishes that last shared a common ancestor with modern jawed vertebrates around 500 million years ago12. Thus, comparisons between lampreys and gnathostomes can identify deeply conserved and evolutionarily flexible features of vertebrate development. Using the frog Xenopus laevis to expand gnathostome phylogenetic representation and facilitate side-by-side analyses, we identify ancient and lineage-specific roles for Edn signalling. These findings suggest that Edn signalling was activated in NCCs before duplication of the vertebrate genome. Then, after one or more genome-wide duplications in the vertebrate stem, paralogous Edn pathways functionally diverged, resulting in NCC subpopulations with different Edn signalling requirements. We posit that this new developmental modularity facilitated the independent evolution of NCC derivatives in stem vertebrates. Consistent with this, differences in Edn pathway targets are associated with differences in the oropharyngeal skeleton and autonomic nervous system of lampreys and modern gnathostomes. In summary, our work provides functional genetic evidence linking the origin and duplication of new vertebrate genes with the stepwise evolution of a defining vertebrate novelty.


Asunto(s)
Endotelinas/metabolismo , Evolución Molecular , Cresta Neural/citología , Petromyzon/metabolismo , Transducción de Señal , Xenopus/metabolismo , Animales , Desarrollo Óseo , Huesos/citología , Huesos/metabolismo , Linaje de la Célula , Endotelinas/genética , Femenino , Cabeza/crecimiento & desarrollo , Corazón/crecimiento & desarrollo , Larva/crecimiento & desarrollo , Ligandos , Masculino , Petromyzon/genética , Petromyzon/crecimiento & desarrollo , Receptores de Endotelina/deficiencia , Receptores de Endotelina/genética , Receptores de Endotelina/metabolismo , Xenopus/genética , Xenopus/crecimiento & desarrollo
10.
Genes Dev ; 32(1): 4-13, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29440224

RESUMEN

The mature heart is composed primarily of four different cell types: cardiac myocytes, endothelium, smooth muscle, and fibroblasts. These cell types derive from pluripotent progenitors that become progressively restricted with regard to lineage potential, giving rise to multipotent cardiac progenitor cells and, ultimately, the differentiated cell types of the heart. Recent studies have begun to shed light on the defining characteristics of the intermediary cell types that exist transiently during this developmental process and the extrinsic and cell-autonomous factors that influence cardiac lineage decisions and cellular competence. This information will shape our understanding of congenital and adult cardiac disease and guide regenerative therapeutic approaches. In addition, cardiac progenitor specification can serve as a model for understanding basic mechanisms regulating the acquisition of cellular identity. In this review, we present the concept of "chromatin competence" that describes the potential for three-dimensional chromatin organization to function as the molecular underpinning of the ability of a progenitor cell to respond to inductive lineage cues and summarize recent studies advancing our understanding of cardiac cell specification, gene regulation, and chromatin organization and how they impact cardiac development.


Asunto(s)
Corazón/crecimiento & desarrollo , Miocardio/citología , Animales , Linaje de la Célula , Cromatina/química , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de Complejo Poro Nuclear/fisiología , Células Madre/citología , Factores de Transcripción/metabolismo
11.
Genes Dev ; 32(19-20): 1344-1357, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30254108

RESUMEN

A fundamental challenge in understanding cardiac biology and disease is that the remarkable heterogeneity in cell type composition and functional states have not been well characterized at single-cell resolution in maturing and diseased mammalian hearts. Massively parallel single-nucleus RNA sequencing (snRNA-seq) has emerged as a powerful tool to address these questions by interrogating the transcriptome of tens of thousands of nuclei isolated from fresh or frozen tissues. snRNA-seq overcomes the technical challenge of isolating intact single cells from complex tissues, including the maturing mammalian hearts; reduces biased recovery of easily dissociated cell types; and minimizes aberrant gene expression during the whole-cell dissociation. Here we applied sNucDrop-seq, a droplet microfluidics-based massively parallel snRNA-seq method, to investigate the transcriptional landscape of postnatal maturing mouse hearts in both healthy and disease states. By profiling the transcriptome of nearly 20,000 nuclei, we identified major and rare cardiac cell types and revealed significant heterogeneity of cardiomyocytes, fibroblasts, and endothelial cells in postnatal developing hearts. When applied to a mouse model of pediatric mitochondrial cardiomyopathy, we uncovered profound cell type-specific modifications of the cardiac transcriptional landscape at single-nucleus resolution, including changes of subtype composition, maturation states, and functional remodeling of each cell type. Furthermore, we employed sNucDrop-seq to decipher the cardiac cell type-specific gene regulatory network (GRN) of GDF15, a heart-derived hormone and clinically important diagnostic biomarker of heart disease. Together, our results present a rich resource for studying cardiac biology and provide new insights into heart disease using an approach broadly applicable to many fields of biomedicine.


Asunto(s)
Perfilación de la Expresión Génica , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Transcriptoma , Animales , Cardiomiopatías/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Redes Reguladoras de Genes , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Enfermedades Mitocondriales/genética , Miocardio/citología , Miocitos Cardíacos/metabolismo , Análisis de Secuencia de ARN , Activación Transcripcional
12.
J Biol Chem ; 300(7): 107434, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38830405

RESUMEN

During postnatal cardiac hypertrophy, cardiomyocytes undergo mitotic exit, relying on DNA replication-independent mechanisms of histone turnover to maintain chromatin organization and gene transcription. In other tissues, circadian oscillations in nucleosome occupancy influence clock-controlled gene expression, suggesting a role for the circadian clock in temporal control of histone turnover and coordinated cardiomyocyte gene expression. We sought to elucidate roles for the master circadian transcription factor, Bmal1, in histone turnover, chromatin organization, and myocyte-specific gene expression and cell growth in the neonatal period. Bmal1 knockdown in neonatal rat ventricular myocytes decreased myocyte size, total cellular protein synthesis, and transcription of the fetal hypertrophic gene Nppb after treatment with serum or the α-adrenergic agonist phenylephrine. Depletion of Bmal1 decreased the expression of clock-controlled genes Per2 and Tcap, as well as Sik1, a Bmal1 target upregulated in adult versus embryonic hearts. Bmal1 knockdown impaired Per2 and Sik1 promoter accessibility as measured by micrococcal nuclease-quantitative PCR and impaired histone turnover as measured by metabolic labeling of acid-soluble chromatin fractions. Sik1 knockdown in turn decreased myocyte size, while simultaneously inhibiting natriuretic peptide B transcription and activating Per2 transcription. Linking these changes to chromatin remodeling, depletion of the replication-independent histone variant H3.3a inhibited myocyte hypertrophy and prevented phenylephrine-induced changes in clock-controlled gene transcription. Bmal1 is required for neonatal myocyte growth, replication-independent histone turnover, and chromatin organization at the Sik1 promoter. Sik1 represents a novel clock-controlled gene that coordinates myocyte growth with hypertrophic and clock-controlled gene transcription. Replication-independent histone turnover is required for transcriptional remodeling of clock-controlled genes in cardiac myocytes in response to growth stimuli.


Asunto(s)
Factores de Transcripción ARNTL , Histonas , Miocitos Cardíacos , Proteínas Circadianas Period , Animales , Histonas/metabolismo , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Ratas , Proteínas Circadianas Period/metabolismo , Proteínas Circadianas Period/genética , Ritmo Circadiano , Fenilefrina/farmacología , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Corazón/embriología , Animales Recién Nacidos , Cardiomegalia/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Ratas Sprague-Dawley , Ensamble y Desensamble de Cromatina , Células Cultivadas , Regiones Promotoras Genéticas
13.
Physiol Rev ; 98(4): 2453-2475, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30156497

RESUMEN

The heart is the first organ to be functional in the fetus. Heart formation is a complex morphogenetic process regulated by both genetic and epigenetic mechanisms. Congenital heart diseases (CHD) are the most prominent congenital diseases. Genetics is not sufficient to explain these diseases or the impact of them on patients. Epigenetics is more and more emerging as a basis for cardiac malformations. This review brings the essential knowledge on cardiac biology of development. It further provides a broad background on epigenetics with a focus on three-dimensional conformation of chromatin. Then, we summarize the current knowledge of the impact of epigenetics on cardiac cell fate decision. We further provide an update on the epigenetic anomalies in the genesis of CHD.


Asunto(s)
Epigénesis Genética/genética , Epigénesis Genética/fisiología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/fisiopatología , Corazón/crecimiento & desarrollo , Animales , Epigenómica/métodos , Feto/fisiología , Humanos
14.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35156682

RESUMEN

The proper development and patterning of organs rely on concerted signaling events emanating from intracellular and extracellular molecular and biophysical cues. The ability to model and understand how these microenvironmental factors contribute to cell fate decisions and physiological processes is crucial for uncovering the biology and mechanisms of life. Recent advances in microfluidic systems have provided novel tools and strategies for studying aspects of human tissue and organ development in ways that have previously been challenging to explore ex vivo. Here, we discuss how microfluidic systems and organs-on-chips provide new ways to understand how extracellular signals affect cell differentiation, how cells interact with each other, and how different tissues and organs are formed for specialized functions. We also highlight key advancements in the field that are contributing to a broad understanding of human embryogenesis, organogenesis and physiology. We conclude by summarizing the key advantages of using dynamic microfluidic or microphysiological platforms to study intricate developmental processes that cannot be accurately modeled by using traditional tissue culture vessels. We also suggest some exciting prospects and potential future applications of these emerging technologies.


Asunto(s)
Microfluídica/métodos , Modelos Biológicos , Corazón/crecimiento & desarrollo , Corazón/fisiología , Humanos , Dispositivos Laboratorio en un Chip , Microfluídica/instrumentación , Poliésteres/química , Impresión Tridimensional , Ingeniería de Tejidos
15.
Development ; 149(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35237793

RESUMEN

The mitochondrial matrix AAA+ Lon protease (LONP1) degrades misfolded or unassembled proteins, which play a pivotal role in mitochondrial quality control. During heart development, a metabolic shift from anaerobic glycolysis to mitochondrial oxidative phosphorylation takes place, which relies strongly on functional mitochondria. However, the relationship between the mitochondrial quality control machinery and metabolic shifts is elusive. Here, we interfered with mitochondrial quality control by inactivating Lonp1 in murine embryonic cardiac tissue, resulting in severely impaired heart development, leading to embryonic lethality. Mitochondrial swelling, cristae loss and abnormal protein aggregates were evident in the mitochondria of Lonp1-deficient cardiomyocytes. Accordingly, the p-eIF2α-ATF4 pathway was triggered, and nuclear translocation of ATF4 was observed. We further demonstrated that ATF4 regulates the expression of Tfam negatively while promoting that of Glut1, which was responsible for the disruption of the metabolic shift to oxidative phosphorylation. In addition, elevated levels of reactive oxygen species were observed in Lonp1-deficient cardiomyocytes. This study revealed that LONP1 safeguards metabolic shifts in the developing heart by controlling mitochondrial protein quality, suggesting that disrupted mitochondrial quality control may cause prenatal cardiomyopathy.


Asunto(s)
Corazón , Mitocondrias Cardíacas , Proteasa La , Proteasas ATP-Dependientes/metabolismo , Animales , Corazón/crecimiento & desarrollo , Ratones , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosforilación Oxidativa , Proteasa La/genética , Proteasa La/metabolismo
18.
J Mol Cell Cardiol ; 191: 50-62, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703412

RESUMEN

Exercise training can promote physiological cardiac growth, which has been suggested to involve changes in glucose metabolism to facilitate hypertrophy of cardiomyocytes. In this study, we used a dietary, in vivo isotope labeling approach to examine how exercise training influences the metabolic fate of carbon derived from dietary glucose in the heart during acute, active, and established phases of exercise-induced cardiac growth. Male and female FVB/NJ mice were subjected to treadmill running for up to 4 weeks and cardiac growth was assessed by gravimetry. Cardiac metabolic responses to exercise were assessed via in vivo tracing of [13C6]-glucose via mass spectrometry and nuclear magnetic resonance. We found that the half-maximal cardiac growth response was achieved by approximately 1 week of daily exercise training, with near maximal growth observed in male mice with 2 weeks of training; however, female mice were recalcitrant to exercise-induced cardiac growth and required a higher daily intensity of exercise training to achieve significant, albeit modest, increases in cardiac mass. We also found that increases in the energy charge of adenylate and guanylate nucleotide pools precede exercise-induced changes in cardiac size and were associated with higher glucose tracer enrichment in the TCA pool and in amino acids (aspartate, glutamate) sourced by TCA intermediates. Our data also indicate that the activity of collateral biosynthetic pathways of glucose metabolism may not be markedly altered by exercise. Overall, this study provides evidence that metabolic remodeling in the form of heightened energy charge and increased TCA cycle activity and cataplerosis precedes cardiac growth caused by exercise training in male mice.


Asunto(s)
Glucosa , Corazón , Miocardio , Condicionamiento Físico Animal , Animales , Masculino , Femenino , Glucosa/metabolismo , Miocardio/metabolismo , Ratones , Corazón/crecimiento & desarrollo , Metabolismo Energético
19.
Am J Physiol Heart Circ Physiol ; 326(6): H1491-H1497, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38668702

RESUMEN

Heart growth in the pregnant patient helps maintain cardiovascular function while supporting the growing fetus. However, in some cases, the cardiovascular demand of pregnancy can trigger life-threatening conditions, including hypertensive disorders of pregnancy and peripartum cardiomyopathy. The mechanisms that control heart growth throughout pregnancy are unclear, and treating these diseases remains elusive. We previously developed a computational model that accounts for hormonal and hemodynamic interactions throughout pregnancy and demonstrated its ability to capture realistic cardiac growth in normal rat pregnancy. In this study, we evaluated whether this model could capture heart growth beyond normal pregnancy. After further validation of our normal pregnancy predictions, we tested our model predictions of three rat studies of hypertensive pregnancies. We next simulated the postpartum period and examined the impact of lactation on cardiac growth in rats. We demonstrate that our multiscale model can capture cardiac growth associated with new-onset hypertension during pregnancy and lactation status in the postpartum period. We conclude by elaborating on the potential clinical utility of our model in the future.NEW & NOTEWORTHY Our multiscale model predicts appropriate heart growth beyond normal pregnancy, including elevated heart weights in rats with induced hypertension during pregnancy and in lactating mice and decreased heart weight in nonlactating mice. Our model captures distinct mechanisms that result in similar organ-level growth, highlighting its potential to distinguish healthy from diseased pregnancy-induced growth.


Asunto(s)
Corazón , Hipertensión Inducida en el Embarazo , Modelos Cardiovasculares , Periodo Posparto , Animales , Femenino , Embarazo , Corazón/fisiopatología , Corazón/crecimiento & desarrollo , Hipertensión Inducida en el Embarazo/fisiopatología , Hipertensión Inducida en el Embarazo/metabolismo , Ratas , Simulación por Computador , Lactancia , Modelos Animales de Enfermedad , Presión Sanguínea , Ratas Sprague-Dawley
20.
Development ; 148(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34027990

RESUMEN

Polycomb repressive complex 2 (PRC2) deposits H3K27me3 on chromatin to silence transcription. PRC2 broadly interacts with RNAs. Currently, the role of the RNA-PRC2 interaction in human cardiogenesis remains elusive. Here, we found that human-specific heart brake lncRNA 1 (HBL1) interacted with two PRC2 subunits, JARID2 and EED, in human pluripotent stem cells (hPSCs). Loss of JARID2, EED or HBL1 significantly enhanced cardiac differentiation from hPSCs. HBL1 depletion disrupted genome-wide PRC2 occupancy and H3K27me3 chromatin modification on essential cardiogenic genes, and broadly enhanced cardiogenic gene transcription in undifferentiated hPSCs and later-on differentiation. In addition, ChIP-seq revealed reduced EED occupancy on 62 overlapped cardiogenic genes in HBL1-/- and JARID2-/- hPSCs, indicating that the epigenetic state of cardiogenic genes was determined by HBL1 and JARID2 at pluripotency stage. Furthermore, after cardiac development occurs, the cytosolic and nuclear fractions of HBL1 could crosstalk via a conserved 'microRNA-1-JARID2' axis to modulate cardiogenic gene transcription. Overall, our findings delineate the indispensable role of HBL1 in guiding PRC2 function during early human cardiogenesis, and expand the mechanistic scope of lncRNA(s) that cytosolic and nuclear portions of HBL1 could coordinate to orchestrate human cardiogenesis.


Asunto(s)
Genoma , Organogénesis , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 2/genética , ARN Largo no Codificante/metabolismo , Diferenciación Celular , Cromatina , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Histonas/genética , Humanos , MicroARNs
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA