Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 11(1): 1064, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32111836

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal and incurable form of interstitial lung disease in which persistent injury results in scar tissue formation. As fibrosis thickens, the lung tissue loses the ability to facilitate gas exchange and provide cells with needed oxygen. Currently, IPF has few treatment options and no effective therapies, aside from lung transplant. Here we present a series of studies utilizing lung spheroid cell-secretome (LSC-Sec) and exosomes (LSC-Exo) by inhalation to treat different models of lung injury and fibrosis. Analysis reveals that LSC-Sec and LSC-Exo treatments could attenuate and resolve bleomycin- and silica-induced fibrosis by reestablishing normal alveolar structure and decreasing both collagen accumulation and myofibroblast proliferation. Additionally, LSC-Sec and LSC-Exo exhibit superior therapeutic benefits than their counterparts derived from mesenchymal stem cells in some measures. We showed that an inhalation treatment of secretome and exosome exhibited therapeutic potential for lung regeneration in two experimental models of pulmonary fibrosis.


Assuntos
Exossomos/transplante , Fibrose Pulmonar Idiopática/terapia , Lesão Pulmonar/terapia , Pulmão/citologia , Esferoides Celulares/metabolismo , Administração por Inalação , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Apoptose/efeitos dos fármacos , Bleomicina/toxicidade , Proliferação de Células , Modelos Animais de Doenças , Exossomos/metabolismo , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Miofibroblastos/citologia , Proteômica , Dióxido de Silício/toxicidade
2.
J Cell Sci ; 132(17)2019 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-31409692

RESUMO

Metastasis accounts for the majority of all cancer deaths, yet the process remains poorly understood. A pivotal step in the metastasis process is the exiting of tumor cells from the circulation, a process known as extravasation. However, it is unclear how tumor cells extravasate and whether multicellular clusters of tumor cells possess the ability to exit as a whole or must first disassociate. In this study, we use in vivo zebrafish and mouse models to elucidate the mechanism tumor cells use to extravasate. We found that circulating tumor cells exit the circulation using the recently identified extravasation mechanism, angiopellosis, and do so as both clusters and individual cells. We further show that when melanoma and cervical cancer cells utilize this extravasation method to exit as clusters, they exhibit an increased ability to form tumors at distant sites through the expression of unique genetic profiles. Collectively, we present a new model for tumor cell extravasation of both individual and multicellular circulating tumor cells.This article has an associated First Person interview with the first author of the paper.


Assuntos
Movimento Celular/fisiologia , Células Neoplásicas Circulantes/metabolismo , Animais , Contagem de Células , Células HeLa , Humanos , Camundongos , Metástase Neoplásica
3.
ACS Nano ; 12(7): 6536-6544, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29943967

RESUMO

Acute liver failure is a critical condition characterized by global hepatocyte death and often time needs a liver transplantation. Such treatment is largely limited by donor organ shortage. Stem cell therapy offers a promising option to patients with acute liver failure. Yet, therapeutic efficacy and feasibility are hindered by delivery route and storage instability of live cell products. We fabricated a nanoparticle that carries the beneficial regenerative factors from mesenchymal stem cells and further coated it with the membranes of red blood cells to increase blood stability. Unlike uncoated nanoparticles, these particles promote liver cell proliferation in vitro and have lower internalization by macrophage cells. After intravenous delivery, these artificial stem cell analogs are able to remain in the liver and mitigate carbon tetrachloride-induced liver failure in a mouse model, as gauged by histology and liver function test. Our technology provides an innovative and off-the-shelf strategy to treat liver failure.


Assuntos
Materiais Biomiméticos/uso terapêutico , Membrana Eritrocítica/química , Falência Hepática Aguda/terapia , Células-Tronco Mesenquimais/química , Nanopartículas/uso terapêutico , Animais , Apoptose , Materiais Biomiméticos/química , Tetracloreto de Carbono , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Humanos , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/patologia , Falência Hepática Aguda/fisiopatologia , Regeneração Hepática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química
4.
ACS Nano ; 11(10): 9738-9749, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-28929735

RESUMO

Stem cell transplantation is currently implemented clinically but is limited by low retention and engraftment of transplanted cells and the adverse effects of inflammation and immunoreaction when allogeneic or xenogeneic cells are used. Here, we demonstrate the safety and efficacy of encapsulating human cardiac stem cells (hCSCs) in thermosensitive poly(N-isopropylacrylamine-co-acrylic acid) or P(NIPAM-AA) nanogel in mouse and pig models of myocardial infarction (MI). Unlike xenogeneic hCSCs injected in saline, injection of nanogel-encapsulated hCSCs does not elicit systemic inflammation or local T cell infiltrations in immunocompetent mice. In mice and pigs with acute MI, injection of encapsulated hCSCs preserves cardiac function and reduces scar sizes, whereas injection of hCSCs in saline has an adverse effect on heart healing. In conclusion, thermosensitive nanogels can be used as a stem cell carrier: the porous and convoluted inner structure allows nutrient, oxygen, and secretion diffusion but can prevent the stem cells from being attacked by immune cells.


Assuntos
Acrilamidas/química , Acrilatos/química , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Polietilenoglicóis/química , Polietilenoimina/química , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Nanogéis , Tamanho da Partícula , Propriedades de Superfície , Suínos , Temperatura
5.
Stem Cells Transl Med ; 6(10): 1905-1916, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28783251

RESUMO

Idiopathic pulmonary fibrosis is a devastating interstitial lung disease characterized by the relentless deposition of extracellular matrix causing lung distortions and dysfunctions. The prognosis after detection is merely 3-5 years and the only two Food and Drug Administration-approved drugs treat the symptoms, not the disease, and have numerous side effects. Stem cell therapy is a promising treatment strategy for pulmonary fibrosis. Current animal and clinical studies focus on the use of adipose or bone marrow-derived mesenchymal stem cells. We, instead, have established adult lung spheroid cells (LSCs) as an intrinsic source of therapeutic lung stem cells. In the present study, we compared the efficacy and safety of syngeneic and allogeneic LSCs in immuno-competent rats with bleomycin-induced pulmonary inflammation in an effort to mitigate fibrosis development. We found that infusion of allogeneic LSCs reduces the progression of inflammation and fibrotic manifestation and preserves epithelial and endothelial health without eliciting significant immune rejection. Our study sheds light on potential future developments of LSCs as an allogeneic cell therapy for humans with pulmonary fibrosis. Stem Cells Translational Medicine 2017;9:1905-1916.


Assuntos
Fibrose Pulmonar/terapia , Esferoides Celulares/transplante , Transplante de Células-Tronco/métodos , Animais , Bleomicina/toxicidade , Células Cultivadas , Feminino , Pulmão/citologia , Fibrose Pulmonar/etiologia , Ratos , Ratos Wistar , Transplante de Células-Tronco/efeitos adversos , Transplante Homólogo/efeitos adversos , Transplante Homólogo/métodos
6.
Respir Res ; 18(1): 132, 2017 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-28666430

RESUMO

BACKGROUND: Resident stem and progenitor cells have been identified in the lung over the last decade, but isolation and culture of these cells remains a challenge. Thus, although these lung stem and progenitor cells provide an ideal source for stem-cell based therapy, mesenchymal stem cells (MSCs) remain the most popular cell therapy product for the treatment of lung diseases. Surgical lung biopsies can be the tissue source but such procedures carry a high risk of mortality. METHODS: In this study we demonstrate that therapeutic lung cells, termed "lung spheroid cells" (LSCs) can be generated from minimally invasive transbronchial lung biopsies using a three-dimensional culture technique. The cells were then characterized by flow cytometry and immunohistochemistry. Angiogenic potential was tested by in-vitro HUVEC tube formation assay. In-vivo bio- distribution of LSCs was examined in athymic nude mice after intravenous delivery. RESULTS: From one lung biopsy, we are able to derive >50 million LSC cells at Passage 2. These cells were characterized by flow cytometry and immunohistochemistry and were shown to represent a mixture of lung stem cells and supporting cells. When introduced systemically into nude mice, LSCs were retained primarily in the lungs for up to 21 days. CONCLUSION: Here, for the first time, we demonstrated that direct culture and expansion of human lung progenitor cells from pulmonary tissues, acquired through a minimally invasive biopsy, is possible and straightforward with a three-dimensional culture technique. These cells could be utilized in long-term expansion of lung progenitor cells and as part of the development of cell-based therapies for the treatment of lung diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF).


Assuntos
Brônquios/citologia , Brônquios/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Esferoides Celulares/fisiologia , Células-Tronco/fisiologia , Adolescente , Idoso , Animais , Biópsia , Técnicas de Cultura de Células/métodos , Feminino , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Infusões Intravenosas , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Células-Tronco/métodos
7.
Stem Cells ; 35(1): 170-180, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27350343

RESUMO

Stem cells possess the ability to home in and travel to damaged tissue when injected intravenously. For the cells to exert their therapeutic effect, they must cross the blood vessel wall and enter the surrounding tissues. The mechanism of extravasation injected stem cells employ for exit has yet to be characterized. Using intravital microscopy and a transgenic zebrafish line Tg(fli1a:egpf) with GFP-expressing vasculature, we documented the detailed extravasation processes in vivo for injected stem cells in comparison to white blood cells (WBCs). While WBCs left the blood vessels by the standard diapedesis process, injected cardiac and mesenchymal stem cells underwent a distinct method of extravasation that was markedly different from diapedesis. Here, the vascular wall undergoes an extensive remodeling to allow the cell to exit the lumen, while the injected cell remains distinctively passive in activity. We termed this process Angio-pello-sis, which represents an alternative mechanism of cell extravasation to the prevailing theory of diapedesis. Stem Cells 2017;35:170-180 Video Highlight: https://youtu.be/i5EI-ZvhBps.


Assuntos
Vasos Sanguíneos/fisiologia , Extravasamento de Materiais Terapêuticos e Diagnósticos/patologia , Animais , Antígenos CD11/metabolismo , Agregação Celular , Membrana Celular/metabolismo , Forma Celular , Cães , Feminino , Humanos , Injeções , Microscopia Intravital , Masculino , Células-Tronco Mesenquimais , Microesferas , Miócitos Cardíacos/citologia , Polímeros/química , Ratos , Fatores de Tempo , Migração Transendotelial e Transepitelial , Peixe-Zebra/metabolismo
8.
Stem Cells Transl Med ; 4(11): 1265-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26359426

RESUMO

UNLABELLED: Lung diseases are devastating conditions and ranked as one of the top five causes of mortality worldwide according to the World Health Organization. Stem cell therapy is a promising strategy for lung regeneration. Previous animal and clinical studies have focused on the use of mesenchymal stem cells (from other parts of the body) for lung regenerative therapies. We report a rapid and robust method to generate therapeutic resident lung progenitors from adult lung tissues. Outgrowth cells from healthy lung tissue explants are self-aggregated into three-dimensional lung spheroids in a suspension culture. Without antigenic sorting, the lung spheroids recapitulate the stem cell niche and contain a natural mixture of lung stem cells and supporting cells. In vitro, lung spheroid cells can be expanded to a large quantity and can form alveoli-like structures and acquire mature lung epithelial phenotypes. In severe combined immunodeficiency mice with bleomycin-induced pulmonary fibrosis, intravenous injection of human lung spheroid cells inhibited apoptosis, fibrosis, and infiltration but promoted angiogenesis. In a syngeneic rat model of pulmonary fibrosis, lung spheroid cells outperformed adipose-derived mesenchymal stem cells in reducing fibrotic thickening and infiltration. Previously, lung spheroid cells (the spheroid model) had only been used to study lung cancer cells. Our data suggest that lung spheroids and lung spheroid cells from healthy lung tissues are excellent sources of regenerative lung cells for therapeutic lung regeneration. SIGNIFICANCE: The results from the present study will lead to future human clinical trials using lung stem cell therapies to treat various incurable lung diseases, including pulmonary fibrosis. The data presented here also provide fundamental knowledge regarding how injected stem cells mediate lung repair in pulmonary fibrosis.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Bleomicina/efeitos adversos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Fibrose Pulmonar/terapia , Regeneração , Esferoides Celulares/transplante , Adulto , Animais , Antibióticos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Feminino , Xenoenxertos , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos SCID , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Esferoides Celulares/metabolismo
9.
Stem Cells Int ; 2015: 960926, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26351465

RESUMO

Despite the efficacy of cardiac stem cells (CSCs) for treatment of cardiomyopathies, there are many limitations to stem cell therapies. CSC-derived exosomes (CSC-XOs) have been shown to be responsible for a large portion of the regenerative effects of CSCs. Using a mouse model of doxorubicin induced dilated cardiomyopathy, we study the effects of systemic delivery of human CSC-XOs in mice. Mice receiving CSC-XOs showed improved heart function via echocardiography, as well as decreased apoptosis and fibrosis. In spite of using immunocompetent mice and human CSC-XOs, mice showed no adverse immune reaction. The use of CSC-XOs holds promise for overcoming the limitations of stem cells and improving cardiac therapies.

10.
Methods Mol Biol ; 1299: 153-60, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25836582

RESUMO

Cardiomyocytes are frequently used for in vitro models for cardiac research. The isolation of cells is time-consuming and, due to the cells limited proliferative abilities, must be performed frequently. To reduce the time requirements and the impact on research animals, we describe a method for cryopreserving neonatal rat cardiomyocytes (NRCMs), and subsequently thawing them for use in assays.


Assuntos
Criopreservação/métodos , Miócitos Cardíacos/citologia , Animais , Animais Recém-Nascidos , Técnicas de Cultura de Células , Células Cultivadas , Ratos
11.
J Am Heart Assoc ; 3(5): e001260, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25300435

RESUMO

BACKGROUND: The regenerative potential of cardiosphere-derived cells (CDCs) for ischemic heart disease has been demonstrated in mice, rats, pigs, and a recently completed clinical trial (CADUCEUS). CDCs are CD105(+) stromal cells of intrinsic cardiac origin, but the antigenic characteristics of the active fraction remain to be defined. CDCs contain a small minority of c-kit(+) cells, which have been argued to be cardiac progenitors, and a variable fraction of CD90(+) cells whose bioactivity is unclear. METHODS: We performed a retrospective analysis of data from the CADUCEUS trial and a prospective mouse study to elucidate the roles of c-kit(+) and CD90(+) cells in human CDCs. Here, we show, surprisingly, that c-kit expression has no relationship to CDCs' therapeutic efficacy in humans, and depletion of c-kit(+) cells does not undermine the structural and functional benefits of CDCs in a mouse model of myocardial infarction (MI). In contrast, CD90 expression negatively correlates with the therapeutic benefit of CDCs in humans (ie, higher CD90 expression associated with lower efficacy). Depletion of CD90(+) cells augments the functional potency of CDCs in murine MI. CD90(-) CDCs secrete lower levels of inflammatory cytokines and can differentiate into cardiomyocytes in vitro and in vivo. CONCLUSION: The majority population of CDCs (CD105(+)/CD90(-)/c-kit(-)) constitutes the active fraction, both in terms of therapeutic efficacy and in the ability to undergo cardiomyogenic differentiation. The c-kit(+) fraction is neither necessary for, nor contributory to, the regenerative efficacy of CDCs.


Assuntos
Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/transplante , Proteínas Proto-Oncogênicas c-kit/metabolismo , Antígenos Thy-1/metabolismo , Análise de Variância , Animais , Apoptose/fisiologia , Biomarcadores/análise , Diferenciação Celular/fisiologia , Transplante de Células/métodos , Células Cultivadas , Modelos Animais de Doenças , Testes de Função Cardíaca , Humanos , Masculino , Camundongos , Camundongos SCID , Infarto do Miocárdio/fisiopatologia , Estudos Prospectivos , Regeneração/fisiologia , Papel (figurativo) , Sensibilidade e Especificidade , Resultado do Tratamento
12.
Nat Commun ; 5: 4880, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25205020

RESUMO

Stem cell transplantation is a promising strategy for therapeutic cardiac regeneration, but current therapies are limited by inefficient interaction between potentially beneficial cells (either exogenously transplanted or endogenously recruited) and the injured tissue. Here we apply targeted nanomedicine to achieve in vivo cell-mediated tissue repair, imaging and localized enrichment without cellular transplantation. Iron nanoparticles are conjugated with two types of antibodies (one against antigens on therapeutic cells and the other directed at injured cells) to produce magnetic bifunctional cell engager (MagBICE). The antibodies link the therapeutic cells to the injured cells, whereas the iron core of MagBICE enables physical enrichment and imaging. We treat acute myocardial infarction by targeting exogenous bone marrow-derived stem cells (expressing CD45) or endogenous CD34-positive cells to injured cardiomyocytes (expressing myosin light chain. Targeting can be further enhanced by magnetic attraction, leading to augmented functional benefits. MagBICE represents a generalizable platform technology for regenerative medicine.


Assuntos
Anticorpos , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Óxido Ferroso-Férrico/uso terapêutico , Leucócitos Mononucleares/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Nanotecnologia/métodos , Células-Tronco , Animais , Movimento Celular , Ferro , Imãs , Ratos , Regeneração
13.
Sci Transl Med ; 3(68): 68ra9, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289273

RESUMO

Autologous or synthetic vascular grafts are used routinely for providing access in hemodialysis or for arterial bypass in patients with cardiovascular disease. However, some patients either lack suitable autologous tissue or cannot receive synthetic grafts. Such patients could benefit from a vascular graft produced by tissue engineering. Here, we engineer vascular grafts using human allogeneic or canine smooth muscle cells grown on a tubular polyglycolic acid scaffold. Cellular material was removed with detergents to render the grafts nonimmunogenic. Mechanical properties of the human vascular grafts were similar to native human blood vessels, and the grafts could withstand long-term storage at 4 °C. Human engineered grafts were tested in a baboon model of arteriovenous access for hemodialysis. Canine grafts were tested in a dog model of peripheral and coronary artery bypass. Grafts demonstrated excellent patency and resisted dilatation, calcification, and intimal hyperplasia. Such tissue-engineered vascular grafts may provide a readily available option for patients without suitable autologous tissue or for those who are not candidates for synthetic grafts.


Assuntos
Prótese Vascular , Engenharia Tecidual/métodos , Enxerto Vascular/métodos , Adolescente , Adulto , Animais , Materiais Biocompatíveis , Cadáver , Células Cultivadas , Cães , Humanos , Masculino , Teste de Materiais , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Papio anubis , Estresse Mecânico , Alicerces Teciduais , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA