Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(15): E3022-E3031, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28348206

RESUMO

Birth defects, which are in part caused by exposure to environmental chemicals and pharmaceutical drugs, affect 1 in every 33 babies born in the United States each year. The current standard to screen drugs that affect embryonic development is based on prenatal animal testing; however, this approach yields low-throughput and limited mechanistic information regarding the biological pathways and potential adverse consequences in humans. To develop a screening platform for molecules that affect human embryonic development based on endothelial cells (ECs) derived from human pluripotent stem cells, we differentiated human pluripotent stem cells into embryonic ECs and induced their maturation under arterial flow conditions. These cells were then used to screen compounds that specifically affect embryonic vasculature. Using this platform, we have identified two compounds that have higher inhibitory effect in embryonic than postnatal ECs. One of them was fluphenazine (an antipsychotic), which inhibits calmodulin kinase II. The other compound was pyrrolopyrimidine (an antiinflammatory agent), which inhibits vascular endothelial growth factor receptor 2 (VEGFR2), decreases EC viability, induces an inflammatory response, and disrupts preformed vascular networks. The vascular effect of the pyrrolopyrimidine was further validated in prenatal vs. adult mouse ECs and in embryonic and adult zebrafish. We developed a platform based on human pluripotent stem cell-derived ECs for drug screening, which may open new avenues of research for the study and modulation of embryonic vasculature.


Assuntos
Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
2.
Small ; 13(15)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28211642

RESUMO

Organ-on-a-chip platforms seek to recapitulate the complex microenvironment of human organs using miniaturized microfluidic devices. Besides modeling healthy organs, these devices have been used to model diseases, yielding new insights into pathophysiology. Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease showing accelerated vascular aging, leading to the death of patients due to cardiovascular diseases. HGPS targets primarily vascular cells, which reside in mechanically active tissues. Here, a progeria-on-a-chip model is developed and the effects of biomechanical strain are examined in the context of vascular aging and disease. Physiological strain induces a contractile phenotype in primary smooth muscle cells (SMCs), while a pathological strain induces a hypertensive phenotype similar to that of angiotensin II treatment. Interestingly, SMCs derived from human induced pluripotent stem cells of HGPS donors (HGPS iPS-SMCs), but not from healthy donors, show an exacerbated inflammatory response to strain. In particular, increased levels of inflammation markers as well as DNA damage are observed. Pharmacological intervention reverses the strain-induced damage by shifting gene expression profile away from inflammation. The progeria-on-a-chip is a relevant platform to study biomechanics in vascular biology, particularly in the setting of vascular disease and aging, while simultaneously facilitating the discovery of new drugs and/or therapeutic targets.


Assuntos
Progressão da Doença , Inflamação/patologia , Dispositivos Lab-On-A-Chip , Progéria/fisiopatologia , Angiotensina II/farmacologia , Fenômenos Biomecânicos , Vasos Sanguíneos/patologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lovastatina/farmacologia , Microfluídica , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fenótipo
3.
Nat Commun ; 15(1): 1816, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418829

RESUMO

The design of human model systems is highly relevant to unveil the underlying mechanisms of aging and to provide insights on potential interventions to extend human health and life span. In this perspective, we explore the potential of 2D or 3D culture models comprising human induced pluripotent stem cells and transdifferentiated cells obtained from aged or age-related disorder-affected donors to enhance our understanding of human aging and to catalyze the discovery of anti-aging interventions.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Idoso , Envelhecimento , Reprogramação Celular/genética , Longevidade
4.
Dev Cell ; 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38897197

RESUMO

Selective degradation of damaged mitochondria by autophagy (mitophagy) is proposed to play an important role in cellular homeostasis. However, the molecular mechanisms and the requirement of mitochondrial quality control by mitophagy for cellular physiology are poorly understood. Here, we demonstrated that primary human cells maintain highly active basal mitophagy initiated by mitochondrial superoxide signaling. Mitophagy was found to be mediated by PINK1/Parkin-dependent pathway involving p62 as a selective autophagy receptor (SAR). Importantly, this pathway was suppressed upon the induction of cellular senescence and in naturally aged cells, leading to a robust shutdown of mitophagy. Inhibition of mitophagy in proliferating cells was sufficient to trigger the senescence program, while reactivation of mitophagy was necessary for the anti-senescence effects of NAD precursors or rapamycin. Furthermore, reactivation of mitophagy by a p62-targeting small molecule rescued markers of cellular aging, which establishes mitochondrial quality control as a promising target for anti-aging interventions.

5.
Nanoscale Horiz ; 6(3): 245-259, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33576750

RESUMO

The vascular bioactivity/safety of nanomaterials is typically evaluated by animal testing, which is of low throughput and does not account for biological differences between animals and humans such as ageing, metabolism and disease profiles. The development of personalized human in vitro platforms to evaluate the interaction of nanomaterials with the vascular system would be important for both therapeutic and regenerative medicine. A library of 30 nanoparticle (NP) formulations, in use in imaging, antimicrobial and pharmaceutical applications, was evaluated in a reporter zebrafish model of vasculogenesis and then tested in personalized humanized models composed of human-induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) with "young" and "aged" phenotypes in 3 vascular network formats: 2D (in polystyrene dish), 3D (in Matrigel) and in a blood vessel on a chip. As a proof of concept, vascular toxicity was used as the main readout. The results show that the toxicity profile of NPs to hiPSC-ECs was dependent on the "age" of the endothelial cells and vascular network format. hiPSC-ECs were less susceptible to the cytotoxicity effect of NPs when cultured in flow than in static conditions, the protective effect being mediated, at least in part, by glycocalyx. Overall, the results presented here highlight the relevance of in vitro hiPSC-derived vascular systems to screen vascular nanomaterial interactions.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Nanopartículas/toxicidade , Adolescente , Animais , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Dispositivos Lab-On-A-Chip , Masculino , Testes de Toxicidade/instrumentação , Testes de Toxicidade/métodos , Peixe-Zebra
6.
Nat Commun ; 11(1): 4110, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807790

RESUMO

Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease in children that leads to early death. Smooth muscle cells (SMCs) are the most affected cells in HGPS individuals, although the reason for such vulnerability remains poorly understood. In this work, we develop a microfluidic chip formed by HGPS-SMCs generated from induced pluripotent stem cells (iPSCs), to study their vulnerability to flow shear stress. HGPS-iPSC SMCs cultured under arterial flow conditions detach from the chip after a few days of culture; this process is mediated by the upregulation of metalloprotease 13 (MMP13). Importantly, double-mutant LmnaG609G/G609GMmp13-/- mice or LmnaG609G/G609GMmp13+/+ mice treated with a MMP inhibitor show lower SMC loss in the aortic arch than controls. MMP13 upregulation appears to be mediated, at least in part, by the upregulation of glycocalyx. Our HGPS-SMCs chip represents a platform for developing treatments for HGPS individuals that may complement previous pre-clinical and clinical treatments.


Assuntos
Metaloproteinase 13 da Matriz/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Biotecnologia/métodos , Doenças Cardiovasculares/metabolismo , Feminino , Frequência Cardíaca/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos , Camundongos Mutantes , Miócitos de Músculo Liso/efeitos dos fármacos , Progéria/metabolismo , Progéria/patologia , Proteômica/métodos
7.
Curr Protoc Stem Cell Biol ; 50(1): e93, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31479593

RESUMO

This article describes a screening platform to identify compounds that affect human embryonic vascular development. The procedure comprises the generation of human embryonic-like endothelial cells (ECs) from human pluripotent stem cells (hPSCs) and subsequent maturation under arterial flow conditions; the use of these cells for the high-throughput screening of small molecules that specifically inhibit the survival of embryonic-like ECs; the confirmation of the hits in embryonic-like ECs cultured under flow shear stress; and final validation in mouse embryonic ECs. The embryonic-like ECs express embryonic genes including DLL1, EPHB2, LYN, TEK, ID1, NRP2, CAST, FLT1, IGF1, DKK3, NIN, LEF1, and SORBS3. The entire screening procedure (without the validation step) can be completed within 1 month. This platform is an alternative/complement to standard animal protocols for assessing the effects of chemicals on embryonic vascular development. © 2019 by John Wiley & Sons, Inc.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Células Cultivadas , Ensaios de Triagem em Larga Escala/métodos , Humanos , Neovascularização Fisiológica
8.
ACS Biomater Sci Eng ; 4(5): 1498-1504, 2018 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-33445307

RESUMO

Aging is characterized by a progressive accumulation of cellular damage, which leads to impaired function. Little is known whether substrates can influence cell aging. This is of utmost importance in the development of medical devices that are in contact with human tissue for long periods of time. To address this question, we have used an accelerated aging cell model derived from Hutchinson-Gilford Progeria Syndrome (HGPS) induced pluripotent stem cells (iPSCs). Our results show that HGPS-iPSC smooth muscle cells (SMCs) have an increased aging profile in substrates with specific micropatterns than in flat ones. This is characterized by an up-regulation in the expression of progerin, ß-galactosidase, annexin 3 and 5, and caspase 9. Signs of cell aging are also observed in SMCs without HGPS cultured in substrates with specific microtopographies. It is further showed that specific micropatterned substrates induce cell aging by triggering a DNA damage program likely by the disruption between cyto- and nucleoskeleton.

9.
Sci Rep ; 6: 34798, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27739443

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare fatal genetic disorder that causes systemic accelerated aging in children. Thanks to the pluripotency and self-renewal properties of induced pluripotent stem cells (iPSC), HGPS iPSC-based modeling opens up the possibility of access to different relevant cell types for pharmacological approaches. In this study, 2800 small molecules were explored using high-throughput screening, looking for compounds that could potentially reduce the alkaline phosphatase activity of HGPS mesenchymal stem cells (MSCs) committed into osteogenic differentiation. Results revealed seven compounds that normalized the osteogenic differentiation process and, among these, all-trans retinoic acid and 13-cis-retinoic acid, that also decreased progerin expression. This study highlights the potential of high-throughput drug screening using HGPS iPS-derived cells, in order to find therapeutic compounds for HGPS and, potentially, for other aging-related disorders.


Assuntos
Senilidade Prematura/terapia , Fosfatase Alcalina/antagonistas & inibidores , Células-Tronco Pluripotentes Induzidas/fisiologia , Isotretinoína/uso terapêutico , Células-Tronco Mesenquimais/fisiologia , Progéria/terapia , Tretinoína/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Criança , Regulação da Expressão Gênica , Regeneração Tecidual Guiada , Ensaios de Triagem em Larga Escala , Humanos , Isotretinoína/farmacologia , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Osteogênese , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA