Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 629(8012): 660-668, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693258

RESUMEN

Ischaemic diseases such as critical limb ischaemia and myocardial infarction affect millions of people worldwide1. Transplanting endothelial cells (ECs) is a promising therapy in vascular medicine, but engrafting ECs typically necessitates co-transplanting perivascular supporting cells such as mesenchymal stromal cells (MSCs), which makes clinical implementation complicated2,3. The mechanisms that enable MSCs to facilitate EC engraftment remain elusive. Here we show that, under cellular stress, MSCs transfer mitochondria to ECs through tunnelling nanotubes, and that blocking this transfer impairs EC engraftment. We devised a strategy to artificially transplant mitochondria, transiently enhancing EC bioenergetics and enabling them to form functional vessels in ischaemic tissues without the support of MSCs. Notably, exogenous mitochondria did not integrate into the endogenous EC mitochondrial pool, but triggered mitophagy after internalization. Transplanted mitochondria co-localized with autophagosomes, and ablation of the PINK1-Parkin pathway negated the enhanced engraftment ability of ECs. Our findings reveal a mechanism that underlies the effects of mitochondrial transfer between mesenchymal and endothelial cells, and offer potential for a new approach for vascular cell therapy.


Asunto(s)
Células Endoteliales , Células Madre Mesenquimatosas , Mitocondrias , Mitofagia , Proteínas Quinasas , Ubiquitina-Proteína Ligasas , Animales , Mitocondrias/metabolismo , Ratones , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/citología , Humanos , Ubiquitina-Proteína Ligasas/metabolismo , Masculino , Proteínas Quinasas/metabolismo , Autofagosomas/metabolismo , Isquemia/metabolismo , Isquemia/terapia , Isquemia/patología , Femenino , Metabolismo Energético , Trasplante de Células Madre Mesenquimatosas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ratones Endogámicos C57BL
2.
Angiogenesis ; 24(2): 327-344, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33454888

RESUMEN

The search for a source of endothelial cells (ECs) with translational therapeutic potential remains crucial in regenerative medicine. Human blood-derived endothelial colony-forming cells (ECFCs) represent a promising source of autologous ECs due to their robust capacity to form vascular networks in vivo and their easy accessibility from peripheral blood. However, whether ECFCs have distinct characteristics with translational value compared to other ECs remains unclear. Here, we show that vascular networks generated with human ECFCs exhibited robust paracrine support for human pluripotent stem cell-derived cardiomyocytes (iCMs), significantly improving protection against drug-induced cardiac injury and enhancing engraftment at ectopic (subcutaneous) and orthotopic (cardiac) sites. In contrast, iCM support was notably absent in grafts with vessels lined by mature-ECs. This differential trophic ability was due to a unique high constitutive expression of the cardioprotective growth factor neuregulin-1 (NRG1). ECFCs, but not mature-ECs, were capable of actively releasing NRG1, which, in turn, reduced apoptosis and increased the proliferation of iCMs via the PI3K/Akt signaling pathway. Transcriptional silencing of NRG1 abrogated these cardioprotective effects. Our study suggests that ECFCs are uniquely suited to support human iCMs, making these progenitor cells ideal for cardiovascular regenerative medicine.


Asunto(s)
Diferenciación Celular , Células Progenitoras Endoteliales/metabolismo , Regulación de la Expresión Génica , Miocitos Cardíacos/metabolismo , Neurregulina-1/biosíntesis , Células Madre Pluripotentes/metabolismo , Células Cultivadas , Humanos , Comunicación Paracrina
3.
Sci Adv ; 6(30): eaba7606, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32832668

RESUMEN

Human induced pluripotent stem cell (h-iPSC)-derived endothelial cells (h-iECs) have become a valuable tool in regenerative medicine. However, current differentiation protocols remain inefficient and lack reliability. Here, we describe a method for rapid, consistent, and highly efficient generation of h-iECs. The protocol entails the delivery of modified mRNA encoding the transcription factor ETV2 at the intermediate mesodermal stage of differentiation. This approach reproducibly differentiated 13 diverse h-iPSC lines into h-iECs with exceedingly high efficiency. In contrast, standard differentiation methods that relied on endogenous ETV2 were inefficient and notably inconsistent. Our h-iECs were functionally competent in many respects, including the ability to form perfused vascular networks in vivo. Timely activation of ETV2 was critical, and bypassing the mesodermal stage produced putative h-iECs with reduced expansion potential and inability to form functional vessels. Our protocol has broad applications and could reliably provide an unlimited number of h-iECs for vascular therapies.


Asunto(s)
Células Endoteliales , Células Madre Pluripotentes Inducidas , Factores de Transcripción , Diferenciación Celular/genética , Células Endoteliales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Blood Adv ; 3(24): 4166-4176, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31851760

RESUMEN

Hemophilia A (HA) is a bleeding disorder caused by mutations in the F8 gene encoding coagulation factor VIII (FVIII). Current treatments are based on regular infusions of FVIII concentrates throughout a patient's life. Alternatively, viral gene therapies that directly deliver F8 in vivo have shown preliminary successes. However, hurdles remain, including lack of infection specificity and the inability to deliver the full-length version of F8 due to restricted viral cargo sizes. Here, we developed an alternative nonviral ex vivo gene-therapy approach that enables the overexpression of full-length F8 in patients' endothelial cells (ECs). We first generated HA patient-specific induced pluripotent stem cells (HA-iPSCs) from urine epithelial cells and genetically modified them using a piggyBac DNA transposon system to insert multiple copies of full-length F8. We subsequently differentiated the modified HA-iPSCs into competent ECs with high efficiency, and demonstrated that the cells (termed HA-FLF8-iECs) were capable of producing high levels of FVIII. Importantly, following subcutaneous implantation into immunodeficient hemophilic (SCID-f8ko) mice, we demonstrated that HA-FLF8-iECs were able to self-assemble into vascular networks, and that the newly formed microvessels had the capacity to deliver functional FVIII directly into the bloodstream of the mice, effectively correcting the clotting deficiency. Moreover, our implant maintains cellular confinement, which reduces potential safety concerns and allows effective monitoring and reversibility. We envision that this proof-of-concept study could become the basis for a novel autologous ex vivo gene-therapy approach to treat HA.


Asunto(s)
Bioingeniería , Factor VIII/genética , Hemofilia A/genética , Hemofilia A/terapia , Microvasos/trasplante , Animales , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Modelos Animales de Enfermedad , Células Madre Embrionarias , Factor VIII/metabolismo , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Células Madre Pluripotentes Inducidas , Ratones , Ratones SCID , Mutación , Fenotipo , Trasplante de Células Madre , Resultado del Tratamiento
5.
Artículo en Inglés | MEDLINE | ID: mdl-28868207

RESUMEN

Notwithstanding remarkable progress in vascular network engineering, implanted bioengineered microvessels largely fail to form anastomoses with the host vasculature. Here, we demonstrate that implants containing assembled human vascular networks (A-Grafts) fail to engraft due to their inability to engage non-inflammatory host neutrophils upon implantation into mice. In contrast, unassembled vascular cells (U-Grafts) readily engage alternatively polarized neutrophils, which in turn serve as indispensable mediators of vascular assembly and anastomosis. The depletion of host neutrophils abrogated vascularization in U-Grafts, whereas an adoptive transfer of neutrophils fully restored vascularization in myeloid-depleted mice. Neutrophil engagement was regulated by secreted factors and was progressively silenced as the vasculature matured. Exogenous addition of factors from U-Grafts reengaged neutrophils and enhanced revascularization in A-Grafts, a process that was recapitulated by blocking Notch signaling. Our data suggest that the pro-vascularization potential of neutrophils can be harnessed to improve the engraftment of bioengineered tissues.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...