Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cell Sci ; 132(17)2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31409692

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Células Neoplásicas Circulantes/metabolismo , Animales , Recuento de Células , Células HeLa , Humanos , Ratones , Metástasis de la Neoplasia
2.
Stem Cells ; 35(1): 170-180, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27350343

RESUMEN

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.


Asunto(s)
Vasos Sanguíneos/fisiología , Extravasación de Materiales Terapéuticos y Diagnósticos/patología , Animales , Antígenos CD11/metabolismo , Agregación Celular , Membrana Celular/metabolismo , Forma de la Célula , Perros , Femenino , Humanos , Inyecciones , Microscopía Intravital , Masculino , Células Madre Mesenquimatosas , Microesferas , Miocitos Cardíacos/citología , Polímeros/química , Ratas , Factores de Tiempo , Migración Transendotelial y Transepitelial , Pez Cebra/metabolismo
3.
Respir Res ; 18(1): 132, 2017 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-28666430

RESUMEN

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).


Asunto(s)
Bronquios/citología , Bronquios/fisiología , Pulmón/citología , Pulmón/fisiología , Esferoides Celulares/fisiología , Células Madre/fisiología , Adolescente , Anciano , Animales , Biopsia , Técnicas de Cultivo de Célula/métodos , Femenino , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Infusiones Intravenosas , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Trasplante de Células Madre/métodos
4.
Theranostics ; 10(8): 3474-3487, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32206102

RESUMEN

Cancer is the second leading cause of death worldwide and patients are in urgent need of therapies that can effectively target cancer with minimal off-target side effects. Exosomes are extracellular nano-shuttles that facilitate intercellular communication between cells and organs. It has been established that tumor-derived exosomes contain a similar protein and lipid composition to that of the cells that secrete them, indicating that exosomes might be uniquely employed as carriers for anti-cancer therapeutics. Methods: We isolated exosomes from two cancer cell lines, then co-cultured each type of cancer cells with these two kinds of exosomes and quantified exosome. HT1080 or Hela exosomes were systemically injected to Nude mice bearing a subcutaneous HT1080 tumor to investigate their cancer-homing behavior. Moreover, cancer cell-derived exosomes were engineered to carry Doxil (a common chemotherapy drug), known as D-exo, were used to detect their target and therapeutic efficacy as anti-cancer drugs. Exosome proteome array analysis were used to reveal the mechanism underly this phenomenon. Results: Exosomes derived from cancer cells fuse preferentially with their parent cancer cells, in vitro. Systemically injected tumor-derived exosomes home to their original tumor tissues. Moreover, compared to Doxil alone, the drug-loaded exosomes showed enhanced therapeutic retention in tumor tissues and eradicated them more effectively in nude mice. Exosome proteome array analysis revealed distinct integrin expression patterns, which might shed light on the underlying mechanisms that explain the exosomal cancer-homing behavior. Conclusion: Here we demonstrate that the exosomes' ability to target the parent cancer is a phenomenon that opens up new ways to devise targeted therapies to deliver anti-tumor drugs.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Exosomas , Animales , Antibióticos Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Exosomas/metabolismo , Células HeLa , Humanos , Ratones , Ratones Desnudos , Polietilenglicoles/administración & dosificación
5.
Nat Commun ; 11(1): 1064, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111836

RESUMEN

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.


Asunto(s)
Exosomas/trasplante , Fibrosis Pulmonar Idiopática/terapia , Lesión Pulmonar/terapia , Pulmón/citología , Esferoides Celulares/metabolismo , Administración por Inhalación , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Bleomicina/toxicidad , Proliferación Celular , Modelos Animales de Enfermedad , Exosomas/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Células Madre Mesenquimatosas/metabolismo , Ratones , Miofibroblastos/citología , Proteómica , Dióxido de Silicio/toxicidad
6.
J Clin Invest ; 129(6): 2237-2250, 2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31033484

RESUMEN

Exosomes, as functional paracrine units of therapeutic cells, can partially reproduce the reparative properties of their parental cells. The constitution of exosomes, as well as their biological activity, largely depends on the cells that secrete them. We isolated exosomes from explant-derived cardiac stromal cells from patients with heart failure (FEXO) or from normal donor hearts (NEXO) and compared their regenerative activities in vitro and in vivo. Patients in the FEXO group exhibited an impaired ability to promote endothelial tube formation and cardiomyocyte proliferation in vitro. Intramyocardial injection of NEXO resulted in structural and functional improvements in a murine model of acute myocardial infarction. In contrast, FEXO therapy exacerbated cardiac function and left ventricular remodeling. microRNA array and PCR analysis revealed dysregulation of miR-21-5p in FEXO. Restoring miR-21-5p expression rescued FEXO's reparative function, whereas blunting miR-21-5p expression in NEXO diminished its therapeutic benefits. Further mechanistic studies revealed that miR-21-5p augmented Akt kinase activity through the inhibition of phosphatase and tensin homolog. Taken together, the heart failure pathological condition altered the miR cargos of cardiac-derived exosomes and impaired their regenerative activities. miR-21-5p contributes to exosome-mediated heart repair by enhancing angiogenesis and cardiomyocyte survival through the phosphatase and tensin homolog/Akt pathway.


Asunto(s)
Exosomas/metabolismo , Insuficiencia Cardíaca/metabolismo , Corazón/fisiología , MicroARNs/metabolismo , Regeneración , Animales , Exosomas/genética , Exosomas/patología , Femenino , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Ratones , MicroARNs/genética , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/patología
7.
Theranostics ; 8(7): 1869-1878, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29556361

RESUMEN

Rationale: Cardiac stem cell-derived exosomes have been demonstrated to promote cardiac regeneration following myocardial infarction in preclinical studies. Recent studies have used intramyocardial injection in order to concentrate exosomes in the infarct. Though effective in a research setting, this method is not clinically appealing due to its invasive nature. We propose the use of a targeting peptide, cardiac homing peptide (CHP), to target intravenously-infused exosomes to the infarcted heart. Methods: Exosomes were conjugated with CHP through a DOPE-NHS linker. Ex vivo targeting was analyzed by incubating organ sections with the CHP exosomes and analyzing with fluorescence microscopy. In vitro assays were performed on neonatal rat cardiomyocytes and H9C2 cells. For the animal study, we utilized an ischemia/reperfusion rat model. Animals were treated with either saline, scramble peptide exosomes, or CHP exosomes 24 h after surgery. Echocardiography was performed 4 h after surgery and 21 d after surgery. At 21 d, animals were sacrificed, and organs were collected for analysis. Results: By conjugating the exosomes with CHP, we demonstrate increased retention of the exosomes within heart sections ex vivo and in vitro with neonatal rat cardiomyocytes. In vitro studies showed improved viability, reduced apoptosis and increased exosome uptake when using CHP-XOs. Using an animal model of ischemia/reperfusion injury, we measured the heart function, infarct size, cellular proliferation, and angiogenesis, with improved outcomes with the CHP exosomes. Conclusions: Our results demonstrate a novel method for increasing delivery of for treatment of myocardial infarction. By targeting exosomes to the infarcted heart, there was a significant improvement in outcomes with reduced fibrosis and scar size, and increased cellular proliferation and angiogenesis.


Asunto(s)
Productos Biológicos/administración & dosificación , Terapia Biológica/métodos , Exosomas , Terapia Molecular Dirigida/métodos , Infarto del Miocardio/terapia , Péptidos/administración & dosificación , Administración Intravenosa , Animales , Productos Biológicos/farmacocinética , Supervivencia Celular , Modelos Animales de Enfermedad , Ecocardiografía , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Péptidos/farmacocinética , Ratas , Resultado del Tratamiento
8.
ACS Appl Mater Interfaces ; 10(39): 33088-33096, 2018 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-30188113

RESUMEN

Cardiac stem cell (CSC) therapy has shown preclinical and clinical evidence for ischemic heart repair but is limited by low cellular engraftment and survival after transplantation. Previous versions of the cardiac patch strategy improve stem cell engraftment and encourage repair of cardiac tissue. However, cardiac patches that can enhance cardiomyogenesis and angiogenesis at the injured site remain elusive. Therapies that target cardiomyocyte proliferation and new blood vessel formation hold great potential for the protection against acute myocardial infarction (MI). Here, we report a new strategy for creating a vascularized cardiac patch in a facile and modular fashion by leveraging microfluidic hydrodynamic focusing to construct the biomimetic microvessels (BMVs) that include human umbilical vein endothelial cells (HUVECs) lining the luminal surface and then encapsulating the BMVs in a fibrin gel spiked with human CSCs. We show that the endothelialized BMVs mimicked the natural architecture and function of capillaries and that the resultant vascularized cardiac patch (BMV-CSC patch) exhibited equivalent release of paracrine factors compared to those of coculture of genuine human CSCs and HUVECs after 7 days of in vitro culture. In a rat model of acute MI, the BMV-CSC patch therapy induced profound mitotic activities of cardiomyocytes in the peri-infarct region 4 weeks post-treatment. A significant increase in myocardial capillary density was noted in the infarcted hearts that received BMV-CSC patch treatment compared to the infarcted hearts treated with conventional CSC patches. The striking therapeutic benefits and the fast and facile fabrication of the BMV-CSC patch make it promising for practical applications. Our findings suggest that the BMV-CSC patch strategy may open up new possibilities for the treatment of ischemic heart injury.


Asunto(s)
Infarto del Miocardio/terapia , Miocardio/citología , Miocitos Cardíacos/citología , Neovascularización Fisiológica/fisiología , Células Madre/citología , Animales , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas Analíticas Microfluídicas , Miocitos Cardíacos/fisiología , Ratas , Ratas Desnudas , Células Madre/fisiología
9.
ACS Nano ; 12(7): 6536-6544, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29943967

RESUMEN

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.


Asunto(s)
Materiales Biomiméticos/uso terapéutico , Membrana Eritrocítica/química , Fallo Hepático Agudo/terapia , Células Madre Mesenquimatosas/química , Nanopartículas/uso terapéutico , Animales , Apoptosis , Materiales Biomiméticos/química , Tetracloruro de Carbono , Línea Celular , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/fisiopatología , Regeneración Hepática , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química
10.
Nat Biomed Eng ; 2: 17-26, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29862136

RESUMEN

Stem cell transplantation, as used clinically, suffers from low retention and engraftment of the transplanted cells. Inspired by the ability of platelets to recruit stem cells to sites of injury on blood vessels, we hypothesized that platelets might enhance the vascular delivery of cardiac stem cells (CSCs) to sites of myocardial infarction injury. Here, we show that CSCs with platelet nanovesicles fused onto their surface membranes express platelet surface markers that are associated with platelet adhesion to injury sites. We also find that the modified CSCs selectively bind collagen-coated surfaces and endothelium-denuded rat aortas, and that in rat and porcine models of acute myocardial infarction the modified CSCs increase retention in the heart and reduce infarct size. Platelet-nanovesicle-fused CSCs thus possess the natural targeting and repairing ability of their parental cell types. This stem cell manipulation approach is fast, straightforward and safe, does not require genetic alteration of the cells, and should be generalizable to multiple cell types.

11.
ACS Nano ; 11(10): 9738-9749, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-28929735

RESUMEN

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.


Asunto(s)
Acrilamidas/química , Acrilatos/química , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Polietilenglicoles/química , Polietileneimina/química , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Nanogeles , Tamaño de la Partícula , Propiedades de Superficie , Porcinos , Temperatura
12.
Tissue Eng Part C Methods ; 23(3): 146-155, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28068869

RESUMEN

Layering a regenerative polymer scaffold on the surface of the heart, termed as a cardiac patch, has been proven to be effective in preserving cardiac function after myocardial infarction (MI). However, the placement of such a patch on the heart usually needs open-chest surgery, which is traumatic, therefore prevents the translation of this strategy into the clinic. We sought to device a way to apply a cardiac patch by spray painting in situ polymerizable biomaterials onto the heart with a minimally invasive procedure. To prove the concept, we used platelet fibrin gel as the "paint" material in a mouse model of MI. The use of the spraying system allowed for placement of a uniform cardiac patch on the heart in a mini-invasive manner without the need for sutures or glue. The spray treatment promoted cardiac repair and attenuated cardiac dysfunction after MI.


Asunto(s)
Plaquetas/metabolismo , Fibrina/metabolismo , Corazón/fisiología , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Regeneración/fisiología , Animales , Materiales Biocompatibles/química , Masculino , Ratones , Infarto del Miocardio/patología , Pinturas , Ratas , Ratas Sprague-Dawley
13.
Nat Commun ; 8: 13724, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045024

RESUMEN

Stem cell therapy represents a promising strategy in regenerative medicine. However, cells need to be carefully preserved and processed before usage. In addition, cell transplantation carries immunogenicity and/or tumourigenicity risks. Mounting lines of evidence indicate that stem cells exert their beneficial effects mainly through secretion (of regenerative factors) and membrane-based cell-cell interaction with the injured cells. Here, we fabricate a synthetic cell-mimicking microparticle (CMMP) that recapitulates stem cell functions in tissue repair. CMMPs carry similar secreted proteins and membranes as genuine cardiac stem cells do. In a mouse model of myocardial infarction, injection of CMMPs leads to the preservation of viable myocardium and augmentation of cardiac functions similar to cardiac stem cell therapy. CMMPs (derived from human cells) do not stimulate T-cell infiltration in immuno-competent mice. In conclusion, CMMPs act as 'synthetic stem cells' which mimic the paracrine and biointerfacing activities of natural stem cells in therapeutic cardiac regeneration.


Asunto(s)
Materiales Biomiméticos/farmacología , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Medios de Cultivo Condicionados/química , Infarto del Miocardio/terapia , Células Madre/metabolismo , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Fraccionamiento Celular , Membrana Celular/química , Membrana Celular/trasplante , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/trasplante , Medios de Cultivo Condicionados/aislamiento & purificación , Modelos Animales de Enfermedad , Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Inyecciones Intralesiones , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Comunicación Paracrina , Recuperación de la Función/efectos de los fármacos , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Stem Cells Transl Med ; 6(10): 1905-1916, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28783251

RESUMEN

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.


Asunto(s)
Fibrosis Pulmonar/terapia , Esferoides Celulares/trasplante , Trasplante de Células Madre/métodos , Animales , Bleomicina/toxicidad , Células Cultivadas , Femenino , Pulmón/citología , Fibrosis Pulmonar/etiología , Ratas , Ratas Wistar , Trasplante de Células Madre/efectos adversos , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/métodos
15.
J Vis Exp ; (98)2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25938862

RESUMEN

Cell culture has become increasingly important in cardiac research, but due to the limited proliferation of cardiomyocytes, culturing cardiomyocytes is difficult and time consuming. The most commonly used cells are neonatal rat cardiomyocytes (NRCMs), which require isolation every time cells are needed. The birth of the rats can be unpredictable. Cryopreservation is proposed to allow for cells to be stored until needed, yet freezing/thawing methods for primary cardiomyocytes are challenging due to the sensitivity of the cells. Using the proper cryoprotectant, dimethyl sulfoxide (DMSO), cryopreservation was achieved. By slowly extracting the DMSO while thawing the cells, cultures were obtained with viable NRCMs. NRCM phenotype was verified using immunocytochemistry staining for α-sarcomeric actinin. In addition, cells also showed spontaneous contraction after several days in culture. Cell viability after thawing was acceptable at 40-60%. In spite of this, the methods outlined allow one to easily cryopreserve and thaw NRCMs. This gives researchers a greater amount of flexibility in planning experiments as well as reducing the use of animals.


Asunto(s)
Criopreservación/métodos , Miocitos Cardíacos/citología , Actinina/análisis , Actinina/metabolismo , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Separación Celular/métodos , Supervivencia Celular/fisiología , Crioprotectores/química , Dimetilsulfóxido/química , Congelación , Miocitos Cardíacos/metabolismo , Ratas , Sarcómeros/química , Sarcómeros/metabolismo
16.
Methods Mol Biol ; 1299: 153-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25836582

RESUMEN

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.


Asunto(s)
Criopreservación/métodos , Miocitos Cardíacos/citología , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Células Cultivadas , Ratas
17.
Stem Cells Transl Med ; 4(11): 1265-74, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26359426

RESUMEN

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.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/terapia , Regeneración , Esferoides Celulares/trasplante , Adulto , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Femenino , Xenoinjertos , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Esferoides Celulares/metabolismo
18.
Stem Cells Int ; 2015: 960926, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26351465

RESUMEN

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.

19.
PLoS One ; 10(11): e0143221, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26599500

RESUMEN

AIMS: The coronary artery ligation model in rodents mimics human myocardial infarction (MI). Normally mechanical ventilation and prolonged anesthesia period are needed. Recently, a method has been developed to create MI by popping-out the heart (without ventilation) followed by immediate suture ligation. Mortality is high due to the time-consuming suture ligation process while the heart is exposed. We sought to improve this method and reduce mortality by rapid coronary ligation using a surgical clip instead of a suture. METHODS AND RESULTS: Mice were randomized into 3 groups: clip MI (CMI), suture MI (SMI), or sham (SHAM). In all groups, heart was manually exposed without intubation through a small incision on the chest wall. Unlike the conventional SMI method, mice in the CMI group received a metal clip on left anterior descending artery (LAD), quickly dispensed by an AutoSuture Surgiclip™. The CMI method took only 1/3 of ligation time of the standard SMI method and improved post-MI survival rate. TTC staining and Masson's trichrome staining revealed a similar degree of infarct size in the SMI and CMI groups. Echocardiograph confirmed that both SMI and CMI groups had a similar reduction of ejection fraction and fraction shortening over the time. Histological analysis showed that the numbers of CD68+ macrophages and apoptotic cells (TUNEL-positive) are indistinguishable between the two groups. CONCLUSION: This new method, taking only less than 3 minutes to complete, represents an efficient myocardial infarction model in rodents.


Asunto(s)
Vasos Coronarios/cirugía , Modelos Animales de Enfermedad , Ligadura , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Instrumentos Quirúrgicos , Animales , Ratones , Infarto del Miocardio/etiología , Infarto del Miocardio/mortalidad , Técnicas de Sutura , Procedimientos Quirúrgicos Vasculares
20.
Biomaterials ; 35(30): 8528-39, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25043570

RESUMEN

Stem cell transplantation is a promising therapeutic strategy for acute or chronic ischemic cardiomyopathy. A major limitation to efficacy in cell transplantation is the low efficiency of retention and engraftment, due at least in part to significant early "wash-out" of cells from coronary blood flow and heart contraction. We sought to enhance cell retention and engraftment by magnetic targeting. Human cardiosphere-derived stem cells (hCDCs) were labeled with FDA-approved ferumoxytol nanoparticles Feraheme(®) (F) in the presence of heparin (H) and protamine (P). FHP labeling is nontoxic to hCDCs. FHP-labeled rat CDCs (FHP-rCDCs) were intracoronarily infused into syngeneic rats, with and without magnetic targeting. Magnetic resonance imaging, fluorescence imaging, and quantitative PCR revealed magnetic targeting increased cardiac retention of transplanted FHP-rCDCs. Neither infusion of FHP-rCDCs nor magnetic targeting exacerbated cardiac inflammation or caused iron overload. The augmentation of acute cell retention translated into more attenuated left ventricular remodeling and greater therapeutic benefit (ejection fraction) 3 weeks after treatment. Histology revealed enhanced cell engraftment and angiogenesis in hearts from the magnetic targeting group. FHP labeling is safe to cardiac stem cells and facilitates magnetically-targeted stem cell delivery into the heart which leads to augmented cell engraftment and therapeutic benefit.


Asunto(s)
Óxido Ferrosoférrico/uso terapéutico , Fenómenos Magnéticos , Infarto del Miocardio/terapia , Miocardio/patología , Nanopartículas/uso terapéutico , Esferoides Celulares/patología , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Óxido Ferrosoférrico/farmacología , Análisis de Elementos Finitos , Pruebas de Función Cardíaca , Heparina/farmacología , Humanos , Inflamación/patología , Sobrecarga de Hierro/patología , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Nanopartículas/ultraestructura , Neovascularización Fisiológica/efectos de los fármacos , Protaminas/farmacología , Ratas , Ratas Endogámicas WKY , Esferoides Celulares/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA