Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 5032, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36977745

RESUMO

Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.


Assuntos
Aterosclerose , Calcinose , Células-Tronco Pluripotentes Induzidas , Progéria , Humanos , Progéria/genética , Everolimo/farmacologia , Everolimo/uso terapêutico , Everolimo/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Aterosclerose/metabolismo , Calcinose/metabolismo , Lamina Tipo A/genética
2.
Stem Cell Reports ; 14(2): 325-337, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32032552

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare disorder caused by a point mutation in the Lamin A gene that produces the protein progerin. Progerin toxicity leads to accelerated aging and death from cardiovascular disease. To elucidate the effects of progerin on endothelial cells, we prepared tissue-engineered blood vessels (viTEBVs) using induced pluripotent stem cell-derived smooth muscle cells (viSMCs) and endothelial cells (viECs) from HGPS patients. HGPS viECs aligned with flow but exhibited reduced flow-responsive gene expression and altered NOS3 levels. Relative to viTEBVs with healthy cells, HGPS viTEBVs showed reduced function and exhibited markers of cardiovascular disease associated with endothelium. HGPS viTEBVs exhibited a reduction in both vasoconstriction and vasodilation. Preparing viTEBVs with HGPS viECs and healthy viSMCs only reduced vasodilation. Furthermore, HGPS viECs produced VCAM1 and E-selectin protein in TEBVs with healthy or HGPS viSMCs. In summary, the viTEBV model has identified a role of the endothelium in HGPS.


Assuntos
Prótese Vascular , Vasos Sanguíneos/fisiopatologia , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Biológicos , Progéria/patologia , Engenharia Tecidual , Vasos Sanguíneos/patologia , Células Clonais , Regulação da Expressão Gênica , Humanos , Masculino , Fenótipo , Doadores de Tecidos
3.
Sci Rep ; 7(1): 8168, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811655

RESUMO

Hutchison-Gilford Progeria Syndrome (HGPS) is a rare, accelerated aging disorder caused by nuclear accumulation of progerin, an altered form of the Lamin A gene. The primary cause of death is cardiovascular disease at about 14 years. Loss and dysfunction of smooth muscle cells (SMCs) in the vasculature may cause defects associated with HGPS. Due to limitations of 2D cell culture and mouse models, there is a need to develop improved models to discover novel therapeutics. To address this need, we produced a functional three-dimensional model of HGPS that replicates an arteriole-scale tissue engineered blood vessel (TEBV) using induced pluripotent stem cell (iPSC)-derived SMCs from an HGPS patient. To isolate the effect of the HGPS iSMCs, the endothelial layer consisted of human cord blood-derived endothelial progenitor cells (hCB-EPCs) from a separate, healthy donor. TEBVs fabricated from HGPS iSMCs and hCB-EPCs show reduced vasoactivity, increased medial wall thickness, increased calcification and apoptosis relative to TEBVs fabricated from normal iSMCs or primary MSCs. Additionally, treatment of HGPS TEBVs with the proposed therapeutic Everolimus, increases HGPS TEBV vasoactivity and increases iSMC differentiation in the TEBVs. These results show the ability of this iPSC-derived TEBV to reproduce key features of HGPS and respond to drugs.


Assuntos
Vasos Sanguíneos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos de Músculo Liso/metabolismo , Progéria/etiologia , Progéria/metabolismo , Engenharia Tecidual , Animais , Biomarcadores , Vasos Sanguíneos/citologia , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Everolimo/farmacologia , Fibroblastos , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Mutação , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Progéria/patologia , Progéria/fisiopatologia , Sirolimo/farmacologia
4.
Biomaterials ; 85: 180-194, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26874281

RESUMO

Access to smooth muscle cells (SMC) would create opportunities for tissue engineering, drug testing, and disease modeling. Herein we report the direct conversion of human endothelial progenitor cells (EPC) to induced smooth muscle cells (iSMC) by induced expression of MYOCD. The EPC undergo a cytoskeletal rearrangement resembling that of mesenchymal cells within 3 days post initiation of MYOCD expression. By day 7, the reprogrammed cells show upregulation of smooth muscle markers ACTA2, MYH11, and TAGLN by qRT-PCR and ACTA2 and MYH11 expression by immunofluorescence. By two weeks, they resemble umbilical artery SMC in microarray gene expression analysis. The iSMC, in contrast to EPC control, show calcium transients in response to phenylephrine stimulation and a contractility an order of magnitude higher than that of EPC as determined by traction force microscopy. Tissue-engineered blood vessels constructed using iSMC show functionality with respect to flow- and drug-mediated vasodilation and vasoconstriction.


Assuntos
Transdiferenciação Celular , Células Progenitoras Endoteliais/citologia , Miócitos de Músculo Liso/citologia , Actinas/genética , Actinas/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Regulação para Baixo , Endoglina/genética , Endoglina/metabolismo , Técnicas de Transferência de Genes , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Engenharia Tecidual , Transativadores/genética , Transativadores/metabolismo , Regulação para Cima
5.
Sci Rep ; 5: 15116, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26456074

RESUMO

Tissue-engineered blood vessels (TEBV) can serve as vascular grafts and may also play an important role in the development of organs-on-a-chip. Most TEBV construction involves scaffolding with biomaterials such as collagen gel or electrospun fibrous mesh. Hypothesizing that a scaffold-free TEBV may be advantageous, we constructed a tubular structure (1 mm i.d.) from aligned human mesenchymal cell sheets (hMSC) as the wall and human endothelial progenitor cell (hEPC) coating as the lumen. The burst pressure of the scaffold-free TEBV was above 200 mmHg after three weeks of sequential culture in a rotating wall bioreactor and perfusion at 6.8 dynes/cm(2). The interwoven organization of the cell layers and extensive extracellular matrix (ECM) formation of the hMSC-based TEBV resembled that of native blood vessels. The TEBV exhibited flow-mediated vasodilation, vasoconstriction after exposure to 1 µM phenylephrine and released nitric oxide in a manner similar to that of porcine femoral vein. HL-60 cells attached to the TEBV lumen after TNF-α activation to suggest a functional endothelium. This study demonstrates the potential of a hEPC endothelialized hMSC-based TEBV for drug screening.


Assuntos
Prótese Vascular , Técnicas de Cultura de Células , Células Endoteliais da Veia Umbilical Humana/citologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Adesão Celular , Matriz Extracelular , Veia Femoral/citologia , Veia Femoral/metabolismo , Células HL-60 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Perfusão , Fenilefrina/farmacologia , Pressão , Suínos , Fator de Necrose Tumoral alfa/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
6.
Artigo em Inglês | MEDLINE | ID: mdl-25570601

RESUMO

Wound healing is a dynamic and complex process of replacing missing or dead cell structures and tissue layers. The aim of this research is to discover biocompatible materials and drugs that can promote cell migration in the wound area and thus enhance desirable wound healing effects. In this paper, we report that PDMS nanogratings could accelerate the migration of epithelial cells along the grating axis, and the addition of Imatinib could further increase the epithelial cell wound healing speed to 1.6 times the speed of control cells. We also demonstrate that this migration is mediated by lamellipodia protrusion, and is Rac1-GTPase activity dependent. Lastly, we discuss the potential application and prospect of different nanostructured biomaterials for wound healing studies.


Assuntos
Movimento Celular , Células Epiteliais/fisiologia , Pseudópodes/fisiologia , Cicatrização , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Meios de Cultura , Cães , Células Madin Darby de Rim Canino , Nanoestruturas/química , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...