Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
2.
Nat Commun ; 15(1): 167, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167821

RESUMEN

Primordial germ cells (PGCs) are the embryonic precursors of sperm and eggs. They transmit genetic and epigenetic information across generations. Given the prominent role of germline defects in diseases such as infertility, detailed understanding of human PGC (hPGC) development has important implications in reproductive medicine and studying human evolution. Yet, hPGC specification remains an elusive process. Here, we report the induction of hPGC-like cells (hPGCLCs) in a bioengineered human pluripotent stem cell (hPSC) culture that mimics peri-implantation human development. In this culture, amniotic ectoderm-like cells (AMLCs), derived from hPSCs, induce hPGCLC specification from hPSCs through paracrine signaling downstream of ISL1. Our data further show functional roles of NODAL, WNT, and BMP signaling in hPGCLC induction. hPGCLCs are successfully derived from eight non-obstructive azoospermia (NOA) participant-derived hPSC lines using this biomimetic platform, demonstrating its promise for screening applications.


Asunto(s)
Células Madre Pluripotentes , Semen , Humanos , Masculino , Células Germinativas/metabolismo , Línea Celular , Transducción de Señal , Diferenciación Celular
3.
Nat Commun ; 14(1): 5435, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37669989

RESUMEN

Cardiogenic growth factors play important roles in heart development. Placental growth factor (PLGF) has previously been reported to have angiogenic effects; however, its potential role in cardiogenesis has not yet been determined. We analyze single-cell RNA-sequencing data derived from human and primate embryonic hearts and find PLGF shows a biphasic expression pattern, as it is expressed specifically on ISL1+ second heart field progenitors at an earlier stage and on vascular smooth muscle cells (SMCs) and endothelial cells (ECs) at later stages. Using chemically modified mRNAs (modRNAs), we generate a panel of cardiogenic growth factors and test their effects on enhancing cardiomyocyte (CM) and EC induction during different stages of human embryonic stem cell (hESC) differentiations. We discover that only the application of PLGF modRNA at early time points of hESC-CM differentiation can increase both CM and EC production. Conversely, genetic deletion of PLGF reduces generation of CMs, SMCs and ECs in vitro. We also confirm in vivo beneficial effects of PLGF modRNA for development of human heart progenitor-derived cardiac muscle grafts on murine kidney capsules. Further, we identify the previously unrecognized PLGF-related transcriptional networks driven by EOMES and SOX17. These results shed light on the dual cardiomyogenic and vasculogenic effects of PLGF during heart development.


Asunto(s)
Células Endoteliales , Miocardio , Femenino , Humanos , Animales , Ratones , Factor de Crecimiento Placentario , Miocitos Cardíacos , Diferenciación Celular
4.
Mol Ther ; 31(3): 866-874, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36528793

RESUMEN

Vascular endothelial growth factor A (VEGF-A) has therapeutic cardiovascular effects, but delivery challenges have impeded clinical development. We report the first clinical study of naked mRNA encoding VEGF-A (AZD8601) injected into the human heart. EPICCURE (ClinicalTrials.gov: NCT03370887) was a randomized, double-blind study of AZD8601 in patients with left ventricular ejection fraction (LVEF) 30%-50% who were undergoing elective coronary artery bypass surgery. Thirty epicardial injections of AZD8601 (total 3 mg) or placebo in citrate-buffered saline were targeted to ischemic but viable myocardial regions mapped using quantitative [15O]-water positron emission tomography. Seven patients received AZD8601 and four received placebo and were followed for 6 months. There were no deaths or treatment-related serious adverse events and no AZD8601-associated infections, immune reactions, or arrhythmias. Exploratory outcomes indicated potential improvement in LVEF, Kansas City Cardiomyopathy Questionnaire scores, and N-terminal pro-B-type natriuretic peptide levels, but the study is limited in size, and significant efficacy conclusions are not possible from the dataset. Naked mRNA without lipid encapsulation may provide a safe delivery platform for introducing genetic material to cardiac muscle, but further studies are needed to confirm efficacy and safety in a larger patient pool.


Asunto(s)
Isquemia Miocárdica , Factor A de Crecimiento Endotelial Vascular , Humanos , Factor A de Crecimiento Endotelial Vascular/genética , Volumen Sistólico , Función Ventricular Izquierda , Puente de Arteria Coronaria/efectos adversos , Puente de Arteria Coronaria/métodos , Corazón , Resultado del Tratamiento , Isquemia Miocárdica/terapia
5.
Mol Ther ; 31(1): 211-229, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-35982619

RESUMEN

Cell-based therapies offer an exciting and novel treatment for heart repair following myocardial infarction (MI). However, these therapies often suffer from poor cell viability and engraftment rates, which involve many factors, including the hypoxic conditions of the infarct environment. Meanwhile, vascular endothelial growth factor (VEGF) has previously been employed as a therapeutic agent to limit myocardial damage and simultaneously induce neovascularization. This study took an approach to transiently overexpress VEGF protein, in a controlled manner, by transfecting human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with VEGF mRNA prior to transplantation. The conditioning of iPSC-CMs with VEGF mRNA ultimately led to greater survival rates of the transplanted cells, which promoted a stable vascular network in the grafted region. Furthermore, bulk RNA transcriptomics data and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) and AGE-RAGE signaling pathways were significantly upregulated in the VEGF-treated iPSC-CMs group. The over-expression of VEGF from iPSC-CMs stimulated cell proliferation and partially attenuated the hypoxic environment in the infarcted area, resulting in reduced ventricular remodeling. This study provides a valuable solution for the survival of transplanted cells in tissue-engineered heart regeneration and may further promote the application of modified mRNA (modRNA) in the field of tissue engineering.


Asunto(s)
Células Madre Pluripotentes Inducidas , Infarto del Miocardio , Trasplante de Células Madre , Factor A de Crecimiento Endotelial Vascular , Animales , Humanos , Ratas , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/cirugía , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Nat Biotechnol ; 40(11): 1586-1600, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36329321

RESUMEN

The extraordinary success of mRNA vaccines against coronavirus disease 2019 (COVID-19) has renewed interest in mRNA as a means of delivering therapeutic proteins. Early clinical trials of mRNA therapeutics include studies of paracrine vascular endothelial growth factor (VEGF) mRNA for heart failure and of CRISPR-Cas9 mRNA for a congenital liver-specific storage disease. However, a series of challenges remains to be addressed before mRNA can be established as a general therapeutic modality with broad relevance to both rare and common diseases. An array of new technologies is being developed to surmount these challenges, including approaches to optimize mRNA cargos, lipid carriers with inherent tissue tropism and in vivo percutaneous delivery systems. The judicious integration of these advances may unlock the promise of biologically targeted mRNA therapeutics, beyond vaccines and other immunostimulatory agents, for the treatment of diverse clinical indications.


Asunto(s)
Vectores Genéticos , ARN Mensajero , Humanos , COVID-19/prevención & control , ARN Mensajero/genética , ARN Mensajero/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vacunas contra la COVID-19
7.
Cell Stem Cell ; 29(9): 1402-1419.e8, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36055194

RESUMEN

Despite its clinical and fundamental importance, our understanding of early human development remains limited. Stem cell-derived, embryo-like structures (or embryoids) allowing studies of early development without using natural embryos can potentially help fill the knowledge gap of human development. Herein, transcriptome at the single-cell level of a human embryoid model was profiled at different time points. Molecular maps of lineage diversifications from the pluripotent human epiblast toward the amniotic ectoderm, primitive streak/mesoderm, and primordial germ cells were constructed and compared with in vivo primate data. The comparative transcriptome analyses reveal a critical role of NODAL signaling in human mesoderm and primordial germ cell specification, which is further functionally validated. Through comparative transcriptome analyses and validations with human blastocysts and in vitro cultured cynomolgus embryos, we further proposed stringent criteria for distinguishing between human blastocyst trophectoderm and early amniotic ectoderm cells.


Asunto(s)
Estratos Germinativos , Análisis de la Célula Individual , Animales , Blastocisto , Linaje de la Célula , Ectodermo , Embrión de Mamíferos , Humanos
8.
Nat Cell Biol ; 24(5): 659-671, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35550611

RESUMEN

Heart regeneration is an unmet clinical need, hampered by limited renewal of adult cardiomyocytes and fibrotic scarring. Pluripotent stem cell-based strategies are emerging, but unravelling cellular dynamics of host-graft crosstalk remains elusive. Here, by combining lineage tracing and single-cell transcriptomics in injured non-human primate heart biomimics, we uncover the coordinated action modes of human progenitor-mediated muscle repair. Chemoattraction via CXCL12/CXCR4 directs cellular migration to injury sites. Activated fibroblast repulsion targets fibrosis by SLIT2/ROBO1 guidance in organizing cytoskeletal dynamics. Ultimately, differentiation and electromechanical integration lead to functional restoration of damaged heart muscle. In vivo transplantation into acutely and chronically injured porcine hearts illustrated CXCR4-dependent homing, de novo formation of heart muscle, scar-volume reduction and prevention of heart failure progression. Concurrent endothelial differentiation contributed to graft neovascularization. Our study demonstrates that inherent developmental programmes within cardiac progenitors are sequentially activated in disease, enabling the cells to sense and counteract acute and chronic injury.


Asunto(s)
Proteínas del Tejido Nervioso , Células Madre Pluripotentes , Animales , Diferenciación Celular , Cicatriz/patología , Cicatriz/prevención & control , Fibrosis , Humanos , Miocardio/patología , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología , Receptores Inmunológicos , Porcinos
9.
Nat Cell Biol ; 24(5): 645-658, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35550612

RESUMEN

The contribution of the epicardium, the outermost layer of the heart, to cardiac regeneration has remained controversial due to a lack of suitable analytical tools. By combining genetic marker-independent lineage-tracing strategies with transcriptional profiling and loss-of-function methods, we report here that the epicardium of the highly regenerative salamander species Pleurodeles waltl has an intrinsic capacity to differentiate into cardiomyocytes. Following cryoinjury, CLDN6+ epicardium-derived cells appear at the lesion site, organize into honeycomb-like structures connected via focal tight junctions and undergo transcriptional reprogramming that results in concomitant differentiation into de novo cardiomyocytes. Ablation of CLDN6+ differentiation intermediates as well as disruption of their tight junctions impairs cardiac regeneration. Salamanders constitute the evolutionarily closest species to mammals with an extensive ability to regenerate heart muscle and our results highlight the epicardium and tight junctions as key targets in efforts to promote cardiac regeneration.


Asunto(s)
Uniones Estrechas , Urodelos , Animales , Mamíferos , Miocardio , Miocitos Cardíacos/patología , Pericardio/patología , Pericardio/fisiología , Urodelos/genética
10.
Nat Commun ; 13(1): 3018, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35641497

RESUMEN

The dysregulated physical interaction between two intracellular membrane proteins, the sarco/endoplasmic reticulum Ca2+ ATPase and its reversible inhibitor phospholamban, induces heart failure by inhibiting calcium cycling. While phospholamban is a bona-fide therapeutic target, approaches to selectively inhibit this protein remain elusive. Here, we report the in vivo application of intracellular acting antibodies (intrabodies), derived from the variable domain of camelid heavy-chain antibodies, to modulate the function of phospholamban. Using a synthetic VHH phage-display library, we identify intrabodies with high affinity and specificity for different conformational states of phospholamban. Rapid phenotypic screening, via modified mRNA transfection of primary cells and tissue, efficiently identifies the intrabody with most desirable features. Adeno-associated virus mediated delivery of this intrabody results in improvement of cardiac performance in a murine heart failure model. Our strategy for generating intrabodies to investigate cardiac disease combined with modified mRNA and adeno-associated virus screening could reveal unique future therapeutic opportunities.


Asunto(s)
Proteínas de Unión al Calcio , Insuficiencia Cardíaca , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Corazón , Ratones , ARN Mensajero
12.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34905512

RESUMEN

Tetralogy of Fallot (TOF) is the most common cyanotic heart defect, yet the underlying genetic mechanisms remain poorly understood. Here, we performed whole-genome sequencing analysis on 146 nonsyndromic TOF parent-offspring trios of Chinese ethnicity. Comparison of de novo variants and recessive genotypes of this data set with data from a European cohort identified both overlapping and potentially novel gene loci and revealed differential functional enrichment between cohorts. To assess the impact of these mutations on early cardiac development, we integrated single-cell and spatial transcriptomics of early human heart development with our genetic findings. We discovered that the candidate gene expression was enriched in the myogenic progenitors of the cardiac outflow tract. Moreover, subsets of the candidate genes were found in specific gene coexpression modules along the cardiomyocyte differentiation trajectory. These integrative functional analyses help dissect the pathogenesis of TOF, revealing cellular hotspots in early heart development resulting in cardiac malformations.


Asunto(s)
Inducción Embrionaria/genética , Corazón/embriología , Tetralogía de Fallot , Pueblo Asiatico/genética , China/epidemiología , Análisis por Conglomerados , Redes Reguladoras de Genes/genética , Estudios de Asociación Genética/métodos , Variación Genética , Humanos , Miocitos Cardíacos/fisiología , Polimorfismo de Nucleótido Simple , Tetralogía de Fallot/etnología , Tetralogía de Fallot/genética , Secuenciación Completa del Genoma/métodos
13.
Mol Med ; 27(1): 102, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496741

RESUMEN

BACKGROUND: The human L39X phospholamban (PLN) cardiomyopathic mutant has previously been reported as a null mutation but the detailed molecular pathways that lead to the complete lack of detectable protein remain to be clarified. Previous studies have shown the implication between an impaired cellular degradation homeostasis and cardiomyopathy development. Therefore, uncovering the underlying mechanism responsible for the lack of PLN protein has important implications in understanding the patient pathology, chronic human calcium dysregulation and aid the development of potential therapeutics. METHODS: A panel of mutant and wild-type reporter tagged PLN modified mRNA (modRNA) constructs were transfected in human embryonic stem cell-derived cardiomyocytes. Lysosomal and proteasomal chemical inhibitors were used together with cell imaging and protein analysis tools in order to dissect degradation pathways associated with expressed PLN constructs. Transcriptional profiling of the cardiomyocytes transfected by wild-type or L39X mutant PLN modRNA was analysed with bulk RNA sequencing. RESULTS: Our modRNA assay system revealed that transfected L39X mRNA was stable and actively translated in vitro but with only trace amount of protein detectable. Proteasomal inhibition of cardiomyocytes transfected with L39X mutant PLN modRNA showed a fourfold increase in protein expression levels. Additionally, RNA sequencing analysis of protein degradational pathways showed a significant distinct transcriptomic signature between wild-type and L39X mutant PLN modRNA transfected cardiomyocytes. CONCLUSION: Our results demonstrate that the cardiomyopathic PLN null mutant L39X is rapidly, actively and specifically degraded by proteasomal pathways. Herein, and to the best of our knowledge, we report for the first time the usage of modified mRNAs to screen for and illuminate alternative molecular pathways found in genes associated with inherited cardiomyopathies.


Asunto(s)
Proteínas de Unión al Calcio/genética , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Homocigoto , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , Alelos , Sustitución de Aminoácidos , Biomarcadores , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/diagnóstico , Línea Celular , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Estabilidad del ARN
14.
Nat Commun ; 12(1): 5126, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446705

RESUMEN

Embryonic development is largely conserved among mammals. However, certain genes show divergent functions. By generating a transcriptional atlas containing >30,000 cells from post-implantation non-human primate embryos, we uncover that ISL1, a gene with a well-established role in cardiogenesis, controls a gene regulatory network in primate amnion. CRISPR/Cas9-targeting of ISL1 results in non-human primate embryos which do not yield viable offspring, demonstrating that ISL1 is critically required in primate embryogenesis. On a cellular level, mutant ISL1 embryos display a failure in mesoderm formation due to reduced BMP4 signaling from the amnion. Via loss of function and rescue studies in human embryonic stem cells we confirm a similar role of ISL1 in human in vitro derived amnion. This study highlights the importance of the amnion as a signaling center during primate mesoderm formation and demonstrates the potential of in vitro primate model systems to dissect the genetics of early human embryonic development.


Asunto(s)
Amnios/metabolismo , Macaca fascicularis/embriología , Mesodermo/embriología , Amnios/embriología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Macaca fascicularis/genética , Macaca fascicularis/metabolismo , Mesodermo/metabolismo , Embarazo , Transducción de Señal
15.
Nat Commun ; 12(1): 5180, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34462437

RESUMEN

Heart failure (HF) is a major cause of morbidity and mortality worldwide, highlighting an urgent need for novel treatment options, despite recent improvements. Aberrant Ca2+ handling is a key feature of HF pathophysiology. Restoring the Ca2+ regulating machinery is an attractive therapeutic strategy supported by genetic and pharmacological proof of concept studies. Here, we study antisense oligonucleotides (ASOs) as a therapeutic modality, interfering with the PLN/SERCA2a interaction by targeting Pln mRNA for downregulation in the heart of murine HF models. Mice harboring the PLN R14del pathogenic variant recapitulate the human dilated cardiomyopathy (DCM) phenotype; subcutaneous administration of PLN-ASO prevents PLN protein aggregation, cardiac dysfunction, and leads to a 3-fold increase in survival rate. In another genetic DCM mouse model, unrelated to PLN (Cspr3/Mlp-/-), PLN-ASO also reverses the HF phenotype. Finally, in rats with myocardial infarction, PLN-ASO treatment prevents progression of left ventricular dilatation and improves left ventricular contractility. Thus, our data establish that antisense inhibition of PLN is an effective strategy in preclinical models of genetic cardiomyopathy as well as ischemia driven HF.


Asunto(s)
Proteínas de Unión al Calcio/genética , Cardiomiopatías/genética , Cardiomiopatías/terapia , Terapia Genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Oligonucleótidos Antisentido/genética , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/metabolismo , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Ratas , Ratas Endogámicas Lew
17.
STAR Protoc ; 2(1): 100339, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33644774

RESUMEN

The combination of population and single-cell RNA sequencing analysis using human embryonic stem cell (hESC) differentiation and developmental tissues is a powerful approach to elucidate an organ-specific cellular and molecular atlas in human embryogenesis. This protocol describes (1) cardiac-directed differentiation and isolation of hESC-derived cardiac derivatives with fluorescence-activated cell sorting, (2) isolation of human embryonic heart-derived single cardiac cells, and (3) construction of cDNA libraries with Smart-seq2. These allow for the preparation of human developmental samples for comprehensive transcriptional analysis. For complete details on the use and execution of this protocol, please refer to Sahara et al. (2019).


Asunto(s)
Diferenciación Celular , Citometría de Flujo , Células Madre Embrionarias Humanas , Miocardio , RNA-Seq , Análisis de la Célula Individual , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Miocardio/citología , Miocardio/metabolismo
20.
Stem Cells ; 38(10): 1267-1278, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32497389

RESUMEN

A family of multipotent heart progenitors plays a central role in the generation of diverse myogenic and nonmyogenic lineages in the heart. Cardiac progenitors in particular play a significant role in lineages involved in disease, and have also emerged to be a strong therapeutic candidate. Based on this premise, we aimed to deeply characterize the progenitor stage of cardiac differentiation at a single-cell resolution. Integrated comparison with an embryonic 5-week human heart transcriptomic dataset validated lineage identities with their late stage in vitro counterparts, highlighting the relevance of an in vitro differentiation for progenitors that are developmentally too early to be accessed in vivo. We utilized trajectory mapping to elucidate progenitor lineage branching points, which are supported by RNA velocity. Nonmyogenic populations, including cardiac fibroblast-like cells and endoderm, were found, and we identified TGFBI as a candidate marker for human cardiac fibroblasts in vivo and in vitro. Both myogenic and nonmyogenic populations express ISL1, and its loss redirected myogenic progenitors into a neural-like fate. Our study provides important insights into processes during early heart development.


Asunto(s)
Linaje de la Célula , Fibroblastos/citología , Células Madre Embrionarias Humanas/citología , Miocardio/citología , Organogénesis , Diferenciación Celular , Linaje de la Célula/genética , Proliferación Celular , Corazón Fetal/fisiología , Fibroblastos/metabolismo , Humanos , Proteínas con Homeodominio LIM/metabolismo , Desarrollo de Músculos , Miocitos Cardíacos/citología , Organogénesis/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...