Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Xenotransplantation ; 30(1): e12786, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36367201

RESUMO

Cardiovascular disease is common and has a high mortality. Due to the limited number of organs available for orthotopic heart transplantation, alternative therapies have received intense interest. In this commentary we contrast xenotransplantation and blastocyst complementation to produce pigs that will serve as donors for organ transplantation. These strategies hold tremendous promise and have the potential to provide an unlimited number of organs for chronic, terminal diseases.


Assuntos
Transplante de Coração , Transplante de Órgãos , Transplantes , Animais , Suínos , Humanos , Transplante Heterólogo , Doadores de Tecidos
2.
J Mol Cell Cardiol ; 172: 109-119, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36030840

RESUMO

End stage heart failure is a terminal disease, and the only curative therapy is orthotopic heart transplantation. Due to limited organ availability, alternative strategies have received intense interest for treatment of patients with advanced heart failure. Recent studies using gene-edited porcine organs suggest that cardiac xenotransplantation may provide a future source of organs. In this review, we highlight the historical milestones for cardiac xenotransplantation and the gene editing strategies designed to overcome immunological barriers, which have culminated in a recent cardiac pig-to-human xenotransplant. We also discuss recent results of studies on the engineering of human-porcine chimeric organs that may provide an alternative and complementary strategy to overcome some of the major immunological barriers to producing a new source of transplantable organs.


Assuntos
Insuficiência Cardíaca , Transplante de Coração , Transplantes , Humanos , Suínos , Animais , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos , Transplante de Coração/métodos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/terapia , Edição de Genes
3.
Am J Transplant ; 22(12): 2786-2790, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36052557

RESUMO

Organ transplantation is limited due to the scarcity of donor organs. In order to expand the supply of organs for transplantation, interspecies chimeras have been examined as a potential future source of humanized organs. Recent studies using gene editing technologies in combination with somatic cell nuclear transfer technology and hiPSCs successfully engineered humanized skeletal muscle in the porcine embryo. As these technologies progress, there are ethical issues that warrant consideration and dialogue.


Assuntos
Células-Tronco Pluripotentes Induzidas , Transplante de Órgãos , Suínos , Animais , Edição de Genes
4.
Arterioscler Thromb Vasc Biol ; 40(12): 2875-2890, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33115267

RESUMO

OBJECTIVE: Endothelial progenitors migrate early during embryogenesis to form the primary vascular plexus. The regulatory mechanisms that govern their migration are not completely defined. Here, we describe a novel role for ETV2 (Ets variant transcription factor 2) in cell migration and provide evidence for an ETV2-Rhoj network as a mechanism responsible for this process. Approach and Results: Analysis of RNAseq datasets showed robust enrichment of migratory/motility pathways following overexpression of ETV2 during mesodermal differentiation. We then analyzed ETV2 chromatin immunoprecipitation-seq and assay for transposase accessible chromatin-seq datasets, which showed enrichment of chromatin immunoprecipitation-seq peaks with increased chromatin accessibility in migratory genes following overexpression of ETV2. Migratory assays showed that overexpression of ETV2 enhanced cell migration in mouse embryonic stem cells, embryoid bodies, and mouse embryonic fibroblasts. Knockout of Etv2 led to migratory defects of Etv2-EYFP+ angioblasts to their predefined regions of developing embryos relative to wild-type controls at embryonic day (E) 8.5, supporting its role during migration. Mechanistically, we showed that ETV2 binds the promoter region of Rhoj serving as an upstream regulator of cell migration. Single-cell RNAseq analysis of Etv2-EYFP+ sorted cells revealed coexpression of Etv2 and Rhoj in endothelial progenitors at E7.75 and E8.25. Overexpression of ETV2 led to a robust increase in Rhoj in both embryoid bodies and mouse embryonic fibroblasts, whereas, its expression was abolished in the Etv2 knockout embryoid bodies. Finally, shRNA-mediated knockdown of Rhoj resulted in migration defects, which were partially rescued by overexpression of ETV2. CONCLUSIONS: These results define an ETV2-Rhoj cascade, which is important for the regulation of endothelial progenitor cell migration.


Assuntos
Movimento Celular , Células-Tronco Embrionárias/enzimologia , Células Progenitoras Endoteliais/enzimologia , Fatores de Transcrição/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Idade Gestacional , Camundongos Transgênicos , Transdução de Sinais , Fatores de Transcrição/genética , Proteínas rho de Ligação ao GTP/genética
5.
J Physiol ; 598(15): 3155-3171, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32406099

RESUMO

KEY POINTS: The decerebrate mouse provides a novel working model of the exercise pressor reflex (EPR). The decerebrate mouse model of the EPR is similar to the previously described decerebrate rat model. Studying the EPR in transgenic mouse models can define exact mechanisms of the EPR in health and disease. ABSTRACT: The exercise pressor reflex (EPR) is defined by a rise in mean arterial pressure (MAP) and heart rate (HR) in response to exercise and is necessary to match metabolic demand and prevent premature fatigue. While this reflex is readily tested in humans, mechanistic studies are largely infeasible. Here, we have developed a novel murine model of the EPR to allow for mechanistic studies in various mouse models. We observed that ventral root stimulation (VRS) in an anaesthetized mouse causes a depressor response and a reduction in HR. In contrast, the same stimulation in a decerebrate mouse causes a rise in MAP and HR which is abolished by dorsal rhizotomy or by neuromuscular blockade. Moreover, we demonstrate a reduced MAP response to VRS using TRPV1 antagonism or in Trpv1 null mice while the response to passive stretch remains intact. Additionally, we demonstrate that intra-arterial infusion of capsaicin results in a dose-related rise in MAP and HR that is significantly reduced by a selective and potent TRPV1 antagonist or is completely abolished in Trpv1 null mice. These data serve to validate the development of a decerebrate mouse model for the study of cardiovascular responses to exercise and further define the role of the TRPV1 receptor in mediating the EPR. This novel model will allow for extensive study of the EPR in unlimited transgenic and mutant mouse lines, and for an unprecedented exploration of the molecular mechanisms that control cardiovascular responses to exercise in health and disease.


Assuntos
Contração Muscular , Reflexo , Animais , Pressão Sanguínea , Estado de Descerebração , Modelos Animais de Doenças , Frequência Cardíaca , Camundongos , Músculo Esquelético , Ratos , Ratos Sprague-Dawley
6.
Reproduction ; 159(5): 539-547, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31990674

RESUMO

Blind enucleation is used in porcine somatic cell nuclear transfer (SCNT) to remove the metaphase II (MII) spindle from the oocyte. Deviation of the MII spindle location, however, leads to incomplete enucleation (IE). Here, we report that the rate of complete enucleation (CE) using the blind method was 80.2 ± 1.7%, although this significantly increased when the polar body-MII deviation was minimized (≦45°). While it is established that IE embryos will not survive to full term, the effect of IE on early stage development is unknown. We have previously demonstrated in mice and pigs that ETV2 deletion results in embryonic lethality due to the lack of hematoendothelial lineages. We observed that ETV2-null cloned embryos derived from blindly and incompletely enucleated oocytes had both WT and mutant sequences at E18 and, using FISH analysis, we observed triploidy. We also compared SCNT embryos generated from either CE or intentionally IE oocytes using the spindle viewer system. We observed a higher in vitro blastocyst rate in the IE versus the CE-SCNT embryos (31.9 ± 3.2% vs 21.0 ± 2.1%). Based on known processes in normal fertilization, we infer that the IE-SCNT embryos extruded the haploid second PB after fusion with donor fibroblasts and formed a near-triploid aneuploid nucleus in each blastomere. These studies demonstrate the peri-implantation survival of residual haploid nuclei following IE and emphasize the need for complete enucleation especially for the analysis of SCNT embryos in the peri-implantation stage and will, further, impact the field of reverse xenotransplantation.


Assuntos
Implantação do Embrião/genética , Desenvolvimento Embrionário/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Clonagem de Organismos/métodos , Técnicas de Cultura Embrionária , Técnicas de Maturação in Vitro de Oócitos , Técnicas de Transferência Nuclear , Suínos , Fatores de Transcrição/metabolismo
8.
J Biol Chem ; 290(15): 9614-25, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25694434

RESUMO

Mesoderm posterior 1 (Mesp1) is well recognized for its role in cardiac development, although it is expressed broadly in mesodermal lineages. We have previously demonstrated important roles for Mesp1 and Ets variant 2 (Etv2) during lineage specification, but their relationship has not been defined. This study reveals that Mesp1 binds to the proximal promoter and transactivates Etv2 gene expression via the CRE motif. We also demonstrate the protein-protein interaction between Mesp1 and cAMP-responsive element binding protein 1 (Creb1) in vitro and in vivo. Utilizing transgenesis, lineage tracing, flow cytometry, and immunostaining technologies, we define the lineage relationship between Mesp1- and Etv2-expressing cell populations. We observe that the majority of Etv2-EYFP(+) cells are derived from Mesp1-Cre(+) cells in both the embryo and yolk sac. Furthermore, we observe that the conditional deletion of Etv2, using a Mesp1-Cre transgenic strategy, results in vascular and hematopoietic defects similar to those observed in the global deletion of Etv2 and that it has embryonic lethality by embryonic day 9.5. In summary, our study supports the hypothesis that Mesp1 is a direct upstream transactivator of Etv2 during embryogenesis and that Creb1 is an important cofactor of Mesp1 in the transcriptional regulation of Etv2 gene expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Fatores de Transcrição/genética , Ativação Transcricional , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Células NIH 3T3 , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
10.
J Card Fail ; 21(9): 761-70, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25934595

RESUMO

Inherited cardiomyopathies, including hypertrophic cardiomyopathy, dilated cardiomyopathies, arrythmogenic right ventricular cardiomyopathy, and other inherited forms of heart failure, represent a unique set of genetically defined cardiovascular disease processes. Unraveling the molecular mechanisms of these deadly forms of human heart disease has been challenging, but recent groundbreaking scientific advances in stem cell technology have allowed for the generation of patient-specific human inducible stem cell (hiPSC)-derived cardiomyocytes (CMs). hiPSC-derived CMs retain the genetic blueprint of the patient, they can be maintained in culture, and they recapitulate the phenotypic characteristics of the disease in vitro, thus serving as a disease in a dish. This review provides an overview of in vitro modeling of inherited cardiomyopathies with the use of patient-specific hiPSC-derived CMs.


Assuntos
Cardiomiopatias , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Cardiovasculares , Miócitos Cardíacos/patologia , Animais , Cardiomiopatias/congênito , Cardiomiopatias/genética , Cardiomiopatias/patologia , Humanos
11.
J Cell Sci ; 125(Pt 22): 5329-37, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22956541

RESUMO

In response to severe injury, adult skeletal muscle exhibits a remarkable regenerative capacity due to a resident muscle stem/progenitor cell population. While a number of factors are expressed in the muscle progenitor cell (MPC) population, the molecular networks that govern this cell population remain an area of active investigation. In this study, utilizing knockdown techniques and overexpression of Foxk1 in the myogenic lineage, we observed dysregulation of Foxo and Mef2 downstream targets. Utilizing an array of technologies, we establish that Foxk1 represses the transcriptional activity of Foxo4 and Mef2 and physically interacts with Foxo4 and Mef2, thus promoting MPC proliferation and antagonizing the myogenic lineage differentiation program, respectively. Correspondingly, knockdown of Foxk1 in C2C12 myoblasts results in cell cycle arrest, and Foxk1 overexpression in C2C12CAR myoblasts retards muscle differentiation. Collectively, we have established that Foxk1 promotes MPC proliferation by repressing Foxo4 transcriptional activity and inhibits myogenic differentiation by repressing Mef2 activity. These studies enhance our understanding of the transcriptional networks that regulate the MPC population and muscle regeneration.


Assuntos
Diferenciação Celular , Fatores de Transcrição Forkhead/metabolismo , Desenvolvimento Muscular , Fatores de Regulação Miogênica/metabolismo , Animais , Ciclo Celular , Proteínas de Ciclo Celular , Proliferação de Células , DNA/metabolismo , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Ligação Proteica , Regeneração , Proteínas Repressoras/metabolismo , Transcrição Gênica
12.
Am J Physiol Heart Circ Physiol ; 306(12): H1610-8, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24727496

RESUMO

Due to their specialized location, stem and progenitor cells are often exposed to oxidative stress. Although ATP-binding cassette transporter subfamily G member 2 (Abcg2)-expressing cells have been implicated in cardiac protective mechanisms involving oxidative stress, there remains a lack of understanding regarding the behavior of cardiac Abcg2-expressing cells when exposed to ROS. The aim of the present study was to characterize the response of the cardiac Abcg2 lineage to oxidative stress. In vitro analysis demonstrated that the antioxidant program regulated by Abcg2 is dependent on a functional transporter. Delivery of paraquat dichloride (PQ), a systemic oxidative stress-inducing agent, to mice confirmed that Abcg2 provides a survival benefit. When exposed to PQ, reporter mice showed an increase in the Abcg2 lineage. Transcriptional and immunohistochemical analysis of Abcg2 lineage-positive cells revealed an enhanced vascular commitment after stress. Finally, preconditioning with PQ demonstrated a reduction in scar size and an increase in angiogenesis after permanent left coronary artery ligation. In conclusion, the data suggest that Abcg2 plays a cytoprotective role in response to in vivo oxidative stress. The contribution of the Abcg2 lineage to the vasculature in the heart is increased after PQ delivery.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Circulação Coronária/fisiologia , Vasos Coronários/fisiologia , Neovascularização Fisiológica/fisiologia , Estresse Oxidativo/fisiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/deficiência , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linhagem da Célula , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Miocárdio/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Paraquat/farmacologia , Espécies Reativas de Oxigênio/farmacologia
13.
Differentiation ; 86(4-5): 184-91, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24361185

RESUMO

BTB-BACK-Kelch (BBK) proteins play broad roles in cellular and molecular regulation. The role of BBK proteins in the skeletal muscle lineage and myogenesis remains an active area of research. Herein, we report a novel BBK gene, Kbtbd5, which we discovered and found to be restricted to the myogenic lineage. We observed that Kbtbd5 was absent in proliferating myoblasts and upregulated upon myogenic differentiation. In situ hybridization analysis revealed that Kbtbd5 was restricted to the skeletal muscle lineage during embryogenesis. We identified a conserved 1.2kb upstream region, which directs reporter expression to the developing skeletal muscle lineage. Transcriptional and mutagenesis assays demonstrated that the E-box motifs contribute to the Kbtbd5 promoter activity. We have also demonstrated the in vivo and in vitro binding between MRFs and the E-box motif in the 1.2kb promoter of the Kbtbd5 gene. Our studies have revealed that the Myod family can transactivate the 1.2kb-luc reporter through the E-box motifs. In addition, we have shown that Kbtbd5 can recruit the Cullin 3 complex in vivo. Using shRNA knockdown, our study has revealed that Kbtbd5 plays an important role in the myogenic differentiation. In summary, we have demonstrated that Kbtbd5 is the direct downstream target gene of the Myod family and regulates myogenic differentiation. Our results further support the notion that Kbtbd5 may serve as an adapter of Cul3 during myogenic differentiation.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína MyoD/genética , Fatores de Transcrição/genética , Animais , Linhagem da Célula , Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Desenvolvimento Muscular/genética , Proteínas Musculares , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/biossíntese
14.
Front Cell Dev Biol ; 11: 1109648, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36923254

RESUMO

Ets variant 2 (Etv2), a member of the Ets factor family, has an essential role in the formation of endothelial and hematopoietic cell lineages during embryonic development. The functional role of ETS transcription factors is, in part, dependent on the interacting proteins. There are relatively few studies exploring the coordinated interplay between ETV2 and its interacting proteins that regulate mesodermal lineage determination. In order to identify novel ETV2 interacting partners, a yeast two-hybrid analysis was performed and the C2H2 zinc finger transcription factor VEZF1 (vascular endothelial zinc finger 1) was identified as a binding factor, which was specifically expressed within the endothelium during vascular development. To confirm this interaction, co-immunoprecipitation and GST pull down assays demonstrated the direct interaction between ETV2 and VEZF1. During embryoid body differentiation, Etv2 achieved its peak expression at day 3.0 followed by rapid downregulation, on the other hand Vezf1 expression increased through day 6 of EB differentiation. We have previously shown that ETV2 potently activated Flt1 gene transcription. Using a Flt1 promoter-luciferase reporter assay, we demonstrated that VEZF1 co-activated the Flt1 promoter. Electrophoretic mobility shift assay and Chromatin immunoprecipitation established VEZF1 binding to the Flt1 promoter. Vezf1 knockout embryonic stem cells had downregulation of hematoendothelial marker genes when undergoing embryoid body mediated mesodermal differentiation whereas overexpression of VEZF1 induced the expression of hematoendothelial genes during differentiation. These current studies provide insight into the co-regulation of the hemato-endothelial lineage development via a co-operative interaction between ETV2 and VEZF1.

15.
Methodist Debakey Cardiovasc J ; 19(5): 16-25, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38028968

RESUMO

Cardiovascular diseases are the number one cause of death worldwide and in the United States (US). Cardiovascular diseases frequently progress to end-stage heart failure, and curative therapies are extremely limited. Intense interest has focused on deciphering the cascades and networks that govern cardiomyocyte proliferation and regeneration of the injured heart. For example, studies have shown that lower organisms such as the adult newt and adult zebrafish have the capacity to completely regenerate their injured heart with restoration of function. Similarly, the neonatal mouse and pig are also able to completely regenerate injured myocardium due to cardiomyocyte proliferation from preexisting cardiomyocytes. Using these animal models and transcriptome analyses, efforts have focused on the definition of factors and signaling pathways that can reactivate and induce cardiomyocyte proliferation in the adult mammalian injured heart. These studies and discoveries have the potential to define novel therapies to promote cardiomyocyte proliferation and repair of the injured, mammalian heart.


Assuntos
Infarto do Miocárdio , Miócitos Cardíacos , Animais , Camundongos , Proliferação de Células , Mamíferos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Suínos , Peixe-Zebra
16.
Cardiovasc Res ; 119(8): 1728-1739, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37036809

RESUMO

AIMS: Congenital heart disease (CHD) is the most common genetic birth defect, which has considerable morbidity and mortality. We focused on deciphering key regulators that govern cardiac progenitors and cardiogenesis. FOXK1 is a forkhead/winged helix transcription factor known to regulate cell cycle kinetics and is restricted to mesodermal progenitors, somites, and heart. In the present study, we define an essential role for FOXK1 during cardiovascular development. METHODS AND RESULTS: We used the mouse embryoid body system to differentiate control and Foxk1 KO embryonic stem cells into mesodermal, cardiac progenitor cells and mature cardiac cells. Using flow cytometry, immunohistochemistry, cardiac beating, transcriptional and chromatin immunoprecipitation quantitative polymerase chain reaction assays, bulk RNA sequencing (RNAseq) and assay for transposase-accessible chromatin using sequencing (ATACseq) analyses, FOXK1 was observed to be an important regulator of cardiogenesis. Flow cytometry analyses revealed perturbed cardiogenesis in Foxk1 KO embryoid bodies (EBs). Bulk RNAseq analysis at two developmental stages showed a significant reduction of the cardiac molecular program in Foxk1 KO EBs compared to the control EBs. ATACseq analysis during EB differentiation demonstrated that the chromatin landscape nearby known important regulators of cardiogenesis was significantly relaxed in control EBs compared to Foxk1 KO EBs. Furthermore, we demonstrated that in the absence of FOXK1, cardiac differentiation was markedly impaired by assaying for cardiac Troponin T expression and cardiac contractility. We demonstrate that FOXK1 is an important regulator of cardiogenesis by repressing the Wnt/ß-catenin signalling pathway and thereby promoting differentiation. CONCLUSION: These results identify FOXK1 as an essential transcriptional and epigenetic regulator of cardiovascular development. Mechanistically, FOXK1 represses Wnt signalling to promote the development of cardiac progenitor cells.


Assuntos
Células-Tronco Embrionárias , Coração , Animais , Camundongos , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Via de Sinalização Wnt
18.
Front Cell Dev Biol ; 10: 1065536, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36568986

RESUMO

The only curative therapy for many endstage diseases is allograft organ transplantation. Due to the limited supply of donor organs, relatively few patients are recipients of a transplanted organ. Therefore, new strategies are warranted to address this unmet need. Using gene editing technologies, somatic cell nuclear transfer and human induced pluripotent stem cell technologies, interspecies chimeric organs have been pursued with promising results. In this review, we highlight the overall technical strategy, the successful early results and the hurdles that need to be addressed in order for these approaches to produce a successful organ that could be transplanted in patients with endstage diseases.

19.
Front Genet ; 13: 948496, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35957684

RESUMO

Focusing on complex extremity trauma and volumetric muscle loss (VML) injuries, this review highlights: 1) the current pathophysiologic limitations of the injury sequela; 2) the gene editing strategy of the pig as a model that provides a novel treatment approach; 3) the notion that human skeletal muscle derived from gene edited, humanized pigs provides a groundbreaking treatment option; and 4) the impact of this technologic platform and how it will advance to far more multifaceted applications. This review seeks to shed insights on a novel treatment option using gene edited pigs as a platform which is necessary to overcome the clinical challenges and limitations in the field.

20.
Nat Cell Biol ; 24(5): 672-684, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35550615

RESUMO

The vasculature is an essential organ for the delivery of blood and oxygen to all tissues of the body and is thus relevant to the treatment of ischaemic diseases, injury-induced regeneration and solid tumour growth. Previously, we demonstrated that ETV2 is an essential transcription factor for the development of cardiac, endothelial and haematopoietic lineages. Here we report that ETV2 functions as a pioneer factor that relaxes closed chromatin and regulates endothelial development. By comparing engineered embryonic stem cell differentiation and reprogramming models with multi-omics techniques, we demonstrated that ETV2 was able to bind nucleosomal DNA and recruit BRG1. BRG1 recruitment remodelled chromatin around endothelial genes and helped to maintain an open configuration, resulting in increased H3K27ac deposition. Collectively, these results will serve as a platform for the development of therapeutic initiatives directed towards cardiovascular diseases and solid tumours.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição , Diferenciação Celular/genética , Cromatina , Nucleossomos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA