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










Base de datos
Intervalo de año de publicación
1.
J Control Release ; 358: 116-127, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37120032

RESUMEN

Several implantable long-acting (LA) delivery systems have been developed for sustained subcutaneous administration of tenofovir alafenamide (TAF), a potent and effective nucleotide reverse transcriptase inhibitor used for HIV pre-exposure prophylaxis (PrEP). LA platforms aim to address the lack of adherence to oral regimens, which has impaired PrEP efficacy. Despite extensive investigations in this field, tissue response to sustained subcutaneous TAF delivery remains to be elucidated as contrasting preclinical results have been reported in the literature. To this end, here we studied the local foreign body response (FBR) to sustained subdermal delivery of three forms of TAF, namely TAF free base (TAFfb), TAF fumarate salt (TAFfs), and TAFfb with urocanic acid (TAF-UA). Sustained constant drug release was achieved via titanium-silicon carbide nanofluidic implants previously shown to be bioinert. The analysis was conducted in both Sprague-Dawley (SD) rats and rhesus macaques over 1.5 and 3 months, respectively. While visual observation did not reveal abnormal adverse tissue reaction at the implantation site, histopathology and Imaging Mass Cytometry (IMC) analyses exposed a local chronic inflammatory response to TAF. In rats, UA mitigated foreign body response to TAF in a concentration-dependent manner. This was not observed in macaques where TAFfb was better tolerated than TAFfs and TAF-UA. Notably, the level of FBR was tightly correlated with local TAF tissue concentration. Further, regardless of the degree of FBR, the fibrotic capsule (FC) surrounding the implants did not interfere with drug diffusion and systemic delivery, as evidenced by TAF PK results and fluorescence recovery after photobleaching (FRAP).


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , Ratas , Animales , Tenofovir , Infecciones por VIH/prevención & control , Macaca mulatta , Ratas Sprague-Dawley , Adenina , Alanina/uso terapéutico
2.
Nat Commun ; 13(1): 7951, 2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36572684

RESUMEN

Pancreatic islet transplantation efficacy for type 1 diabetes (T1D) management is limited by hypoxia-related graft attrition and need for systemic immunosuppression. To overcome these challenges, we developed the Neovascularized Implantable Cell Homing and Encapsulation (NICHE) device, which integrates direct vascularization for facile mass transfer and localized immunosuppressant delivery for islet rejection prophylaxis. Here, we investigated NICHE efficacy for allogeneic islet transplantation and long-term diabetes reversal in an immunocompetent, male rat model. We demonstrated that allogeneic islets transplanted within pre-vascularized NICHE were engrafted, revascularized, and functional, reverting diabetes in rats for over 150 days. Notably, we confirmed that localized immunosuppression prevented islet rejection without inducing toxicity or systemic immunosuppression. Moreover, for translatability efforts, we showed NICHE biocompatibility and feasibility of deployment as well as short-term allogeneic islet engraftment in an MHC-mismatched nonhuman primate model. In sum, the NICHE holds promise as a viable approach for safe and effective islet transplantation and long-term T1D management.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Ratas , Animales , Masculino , Diabetes Mellitus Tipo 1/terapia , Terapia de Inmunosupresión , Tolerancia Inmunológica , Inmunosupresores/farmacología , Supervivencia de Injerto
3.
Nanomedicine ; 36: 102419, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34147665

RESUMEN

In this study we produced a set of in vitro culture platforms to model vascular cell responses to growth factors and factor delivery vehicles. Two of the systems (whole vessel and whole lung vascular development) were supported by microfluidic systems facilitating media circulation and waste removal. We assessed vascular endothelial growth factor (VEGF) delivery by Pluronic F-127 hydrogel, 30 nm pore-sized microparticles (MPs), 60 nm pore-sized MP or a 50/50 mixture of 30 and 60 nm pore-sized MP. VEGF was delivered to porcine acellular lung vascular scaffolds (2.5 cm2 square pieces or whole 3D segments of acellular blood vessels) as well as whole acellular lung scaffolds. Scaffold-cell attachment was examined as was vascular tissue formation. We showed that a 50/50 mixture of 30 and 60 nm pore-sized silicon wafer MPs allowed for long-term release of VEGF within the scaffold vasculature and supported vascular endothelial tissue development during in vitro culture.


Asunto(s)
Portadores de Fármacos , Células Endoteliales/metabolismo , Hidrogeles , Pulmón , Neovascularización Fisiológica/efectos de los fármacos , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular , Animales , Técnicas de Cultivo de Célula , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Hidrogeles/química , Hidrogeles/farmacocinética , Hidrogeles/farmacología , Pulmón/irrigación sanguínea , Pulmón/química , Porosidad , Porcinos , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacocinética , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
Adv Ther (Weinh) ; 4(3)2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33997267

RESUMEN

Pre-exposure prophylaxis (PrEP) using antiretroviral oral drugs is effective at preventing HIV transmission when individuals adhere to the dosing regimen. Tenofovir alafenamide (TAF) is a potent antiretroviral drug, with numerous long-acting (LA) delivery systems under development to improve PrEP adherence. However, none has undergone preventive efficacy assessment. Here we show that LA TAF using a novel subcutaneous nanofluidic implant (nTAF) confers partial protection from HIV transmission. We demonstrate that sustained subcutaneous delivery through nTAF in rhesus macaques maintained tenofovir diphosphate concentration at a median of 390.00 fmol/106 peripheral blood mononuclear cells, 9 times above clinically protective levels. In a non-blinded, placebo-controlled rhesus macaque study with repeated low-dose rectal SHIVSF162P3 challenge, the nTAF cohort had a 62.50% reduction (95% CI: 1.72% to 85.69%; p=0.068) in risk of infection per exposure compared to the control. Our finding mirrors that of tenofovir disoproxil fumarate (TDF) monotherapy, where 60.00% protective efficacy was observed in macaques, and clinically, 67.00% reduction in risk with 86.00% preventive efficacy in individuals with detectable drug in the plasma. Overall, our nanofluidic technology shows potential as a subcutaneous delivery platform for long-term PrEP and provides insights for clinical implementation of LA TAF for HIV prevention.

5.
Adv Healthc Mater ; 9(19): e2000670, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32864893

RESUMEN

The current standard for cell encapsulation platforms is enveloping cells in semipermeable membranes that physically isolate transplanted cells from the host while allowing for oxygen and nutrient diffusion. However, long-term viability and function of encapsulated cells are compromised by insufficient oxygen and nutrient supply to the graft. To address this need, a strategy to achieve enhanced vascularization of a 3D-printed, polymeric cell encapsulation platform using platelet-rich plasma (PRP) and mesenchymal stem cells (MSCs) is investigated. The study is conducted in rats and, for clinical translation relevance, in nonhuman primates (NHP). Devices filled with PRP, MSCs, or vehicle hydrogel are subcutaneously implanted in rats and NHP and the amount and maturity of penetrating blood vessels assessed via histopathological analysis. In rats, MSCs drive the strongest angiogenic response at early time points, with the highest vessel density and endothelial nitric oxide synthase (eNOS) expression. In NHP, PRP and MSCs result in similar vessel densities but incorporation of PRP ensues higher levels of eNOS expression. Overall, enrichment with PRP and MSCs yields extensive, mature vascularization of subcutaneous cell encapsulation devices. It is postulated that the individual properties of PRP and MSCs can be leveraged in a synergistic approach for maximal vascularization of cell encapsulation platforms.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Plasma Rico en Plaquetas , Animales , Encapsulación Celular , Hidrogeles , Impresión Tridimensional , Ratas
6.
Biomaterials ; 257: 120232, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32768727

RESUMEN

Cell encapsulation is an attractive transplantation strategy to treat endocrine disorders. Transplanted cells offer a dynamic and stimulus-responsive system that secretes therapeutics based on patient need. Despite significant advancements, a challenge in allogeneic cell encapsulation is maintaining sufficient oxygen and nutrient exchange, while providing protection from the host immune system. To this end, we developed a subcutaneously implantable dual-reservoir encapsulation system integrating in situ prevascularization and local immunosuppressant delivery, termed NICHE. NICHE structure is 3D-printed in biocompatible polyamide 2200 and comprises of independent cell and drug reservoirs separated by a nanoporous membrane for sustained local release of immunosuppressant. Here we present the development and characterization of NICHE, as well as efficacy validation for allogeneic cell transplantation in an immunocompetent rat model. We established biocompatibility and mechanical stability of NICHE. Further, NICHE vascularization was achieved with the aid of mesenchymal stem cells. Our study demonstrated sustained local elution of immunosuppressant (CTLA4Ig) into the cell reservoir protected transcutaneously-transplanted allogeneic Leydig cells from host immune destruction during a 31-day study, and reduced systemic drug exposure by 12-fold. In summary, NICHE is the first encapsulation platform achieving both in situ vascularization and immunosuppressant delivery, presenting a viable strategy for allogeneic cell transplantation.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Preparaciones Farmacéuticas , Animales , Encapsulación Celular , Inmunosupresores , Masculino , Ratas , Trasplante Homólogo
7.
Virology ; 534: 80-86, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31220651

RESUMEN

The immunopathological mechanisms as well as the role played by influenza A virus infection of human leukocytes and induction of apoptosis have not been fully elucidated. We confirm here that the percentage of cells that are infected is less than the percent of apoptotic cells. Depletion of monocytes/macrophages and depletion of cells expressing influenza neuraminidase from the cultures after exposure to virus decreased lymphocyte apoptosis. Treatment of virus-exposed leukocyte cultures with anti-neuraminidase antibodies but not with anti-hemagglutinin antibodies, reduced lymphocyte production of active caspase-3 and induction of apoptosis. Different strains of virus induced different levels of apoptosis. Variations in induction of apoptosis correlated with production and expression of viral neuraminidase by infected leukocytes. The data suggest that cell surface expression of neuraminidase plays an important role in the induction of apoptosis in human lymphocytes. The benefit, or cost, to the host of lymphocyte apoptosis warrants continued investigation.


Asunto(s)
Apoptosis , Membrana Celular/virología , Virus de la Influenza A/enzimología , Gripe Humana/virología , Linfocitos/citología , Neuraminidasa/metabolismo , Proteínas Virales/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Gripe Humana/enzimología , Gripe Humana/genética , Gripe Humana/fisiopatología , Linfocitos/virología , Neuraminidasa/genética , Proteínas Virales/genética
8.
Sci Transl Med ; 10(452)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068570

RESUMEN

The inability to produce perfusable microvasculature networks capable of supporting tissue survival and of withstanding physiological pressures without leakage is a fundamental problem facing the field of tissue engineering. Microvasculature is critically important for production of bioengineered lung (BEL), which requires systemic circulation to support tissue survival and coordination of circulatory and respiratory systems to ensure proper gas exchange. To advance our understanding of vascularization after bioengineered organ transplantation, we produced and transplanted BEL without creation of a pulmonary artery anastomosis in a porcine model. A single pneumonectomy, performed 1 month before BEL implantation, provided the source of autologous cells used to bioengineer the organ on an acellular lung scaffold. During 30 days of bioreactor culture, we facilitated systemic vessel development using growth factor-loaded microparticles. We evaluated recipient survival, autograft (BEL) vascular and parenchymal tissue development, graft rejection, and microbiome reestablishment in autografted animals 10 hours, 2 weeks, 1 month, and 2 months after transplant. BEL became well vascularized as early as 2 weeks after transplant, and formation of alveolar tissue was observed in all animals (n = 4). There was no indication of transplant rejection. BEL continued to develop after transplant and did not require addition of exogenous growth factors to drive cell proliferation or lung and vascular tissue development. The sterile BEL was seeded and colonized by the bacterial community of the native lung.


Asunto(s)
Ingeniería Biomédica , Trasplante de Pulmón , Animales , Regulación de la Expresión Génica , Inmunidad , Pulmón/crecimiento & desarrollo , Pulmón/inmunología , Pulmón/ultraestructura , Linfangiogénesis/genética , Microbiota , Modelos Animales , Porcinos , Andamios del Tejido/química , Transcriptoma/genética
9.
Biomaterials ; 177: 125-138, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29886385

RESUMEN

Autologous cell transplantation holds enormous promise to restore organ and tissue functions in the treatment of various pathologies including endocrine, cardiovascular, and neurological diseases among others. Even though immune rejection is circumvented with autologous transplantation, clinical adoption remains limited due to poor cell retention and survival. Cell transplant success requires homing to vascularized environment, cell engraftment and importantly, maintenance of inherent cell function. To address this need, we developed a three dimensional (3D) printed cell encapsulation device created with polylactic acid (PLA), termed neovascularized implantable cell homing and encapsulation (NICHE). In this paper, we present the development and systematic evaluation of the NICHE in vitro, and the in vivo validation with encapsulated testosterone-secreting Leydig cells in Rag1-/- castrated mice. Enhanced subcutaneous vascularization of NICHE via platelet-rich plasma (PRP) hydrogel coating and filling was demonstrated in vivo via a chorioallantoic membrane (CAM) assay as well as in mice. After establishment of a pre-vascularized bed within the NICHE, transcutaneously transplanted Leydig cells, maintained viability and robust testosterone secretion for the duration of the study. Immunohistochemical analysis revealed extensive Leydig cell colonization in the NICHE. Furthermore, transplanted cells achieved physiologic testosterone levels in castrated mice. The promising results provide a proof of concept for the NICHE as a viable platform technology for autologous cell transplantation for the treatment of a variety of diseases.


Asunto(s)
Materiales Biocompatibles/química , Células Intersticiales del Testículo/trasplante , Poliésteres/química , Andamios del Tejido/química , Animales , Supervivencia Celular , Células Cultivadas , Células Inmovilizadas/citología , Células Inmovilizadas/trasplante , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Islotes Pancreáticos/citología , Células Intersticiales del Testículo/citología , Masculino , Ratones , Neovascularización Fisiológica , Impresión Tridimensional , Ingeniería de Tejidos
10.
J Tissue Eng Regen Med ; 11(7): 2136-2152, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-26756722

RESUMEN

We report, for the first time, the development of an organ culture system and protocols to support recellularization of whole acellular (AC) human paediatric lung scaffolds. The protocol for paediatric lung recellularization was developed using human transformed or immortalized cell lines and single human AC lung scaffolds. Using these surrogate cell populations, we identified cell number requirements, cell type and order of cell installations, flow rates and bioreactor management methods necessary for bioengineering whole lungs. Following the development of appropriate cell installation protocols, paediatric AC scaffolds were recellularized using primary lung alveolar epithelial cells (AECs), vascular cells and tracheal/bronchial cells isolated from discarded human adult lungs. Bioengineered paediatric lungs were shown to contain well-developed vascular, respiratory epithelial and lung tissue, with evidence of alveolar-capillary junction formation. Types I and II AECs were found thoughout the paediatric lungs. Furthermore, surfactant protein-C and -D and collagen I were produced in the bioengineered lungs, which resulted in normal lung compliance measurements. Although this is a first step in the process of developing tissues for transplantation, this study demonstrates the feasibility of producing bioengineered lungs for clinical use. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Bioprótesis , Reactores Biológicos , Pulmón/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Células Epiteliales Alveolares/citología , Animales , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino
11.
Exp Biol Med (Maywood) ; 239(9): 1135-69, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24962174

RESUMEN

Respiratory tract specific cell populations, or tissue engineered in vitro grown human lung, have the potential to be used as research tools to mimic physiology, toxicology, pathology, as well as infectious diseases responses of cells or tissues. Studies related to respiratory tract pathogenesis or drug toxicity testing in the past made use of basic systems where single cell populations were exposed to test agents followed by evaluations of simple cellular responses. Although these simple single-cell-type systems provided good basic information related to cellular responses, much more can be learned from cells grown in fabricated microenvironments which mimic in vivo conditions in specialized microfabricated chambers or by human tissue engineered three-dimensional (3D) models which allow for more natural interactions between cells. Recent advances in microengineering technology, microfluidics, and tissue engineering have provided a new approach to the development of 2D and 3D cell culture models which enable production of more robust human in vitro respiratory tract models. Complex models containing multiple cell phenotypes also provide a more reasonable approximation of what occurs in vivo without the confounding elements in the dynamic in vivo environment. The goal of engineering good 3D human models is the formation of physiologically functional respiratory tissue surrogates which can be used as pathogenesis models or in the case of 2D screening systems for drug therapy evaluation as well as human toxicity testing. We hope that this manuscript will serve as a guide for development of future respiratory tract model systems as well as a review of conventional models.


Asunto(s)
Pulmón , Modelos Biológicos , Técnicas de Cultivo de Tejidos , Ingeniería de Tejidos , Animales , Humanos , Pulmón/citología , Pulmón/metabolismo , Técnicas de Cultivo de Tejidos/instrumentación , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
12.
Tissue Eng Part A ; 19(17-18): 2045-62, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23638920

RESUMEN

The authors have previously shown that acellular (AC) trachea-lung scaffolds can (1) be produced from natural rat lungs, (2) retain critical components of the extracellular matrix (ECM) such as collagen-1 and elastin, and (3) be used to produce lung tissue after recellularization with murine embryonic stem cells. The aim of this study was to produce large (porcine or human) AC lung scaffolds to determine the feasibility of producing scaffolds with potential clinical applicability. We report here the first attempt to produce AC pig or human trachea-lung scaffold. Using a combination of freezing and sodium dodecyl sulfate washes, pig trachea-lungs and human trachea-lungs were decellularized. Once decellularization was complete we evaluated the structural integrity of the AC lung scaffolds using bronchoscopy, multiphoton microscopy (MPM), assessment of the ECM utilizing immunocytochemistry and evaluation of mechanics through the use of pulmonary function tests (PFTs). Immunocytochemistry indicated that there was loss of collagen type IV and laminin in the AC lung scaffold, but retention of collagen-1, elastin, and fibronectin in some regions. MPM scoring was also used to examine the AC lung scaffold ECM structure and to evaluate the amount of collagen I in normal and AC lung. MPM was used to examine the physical arrangement of collagen-1 and elastin in the pleura, distal lung, lung borders, and trachea or bronchi. MPM and bronchoscopy of trachea and lung tissues showed that no cells or cell debris remained in the AC scaffolds. PFT measurements of the trachea-lungs showed no relevant differences in peak pressure, dynamic or static compliance, and a nonrestricted flow pattern in AC compared to normal lungs. Although there were changes in content of collagen I and elastin this did not affect the mechanics of lung function as evidenced by normal PFT values. When repopulated with a variety of stem or adult cells including human adult primary alveolar epithelial type II cells both pig and human AC scaffolds supported cell attachment and cell viability. Examination of scaffolds produced using a variety of detergents indicated that detergent choice influenced human immune response in terms of T cell activation and chemokine production.


Asunto(s)
Pulmón , Andamios del Tejido/química , Animales , Colágeno/química , Humanos , Inmunohistoquímica , Laminina/química , Porcinos , Ingeniería de Tejidos/métodos
13.
Stem Cell Res Ther ; 4(1): 3, 2013 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-23290300

RESUMEN

INTRODUCTION: Nervous system injuries comprise a diverse group of disorders that include traumatic brain injury (TBI). The potential of mesenchymal stem cells (MSCs) to differentiate into neural cell types has aroused hope for the possible development of autologous therapies for central nervous system injury. METHODS: In this study we isolated and characterized a human peripheral blood derived (HPBD) MSC population which we examined for neural lineage potential and ability to migrate in vitro and in vivo. HPBD CD133+, ATP-binding cassette sub-family G member 2 (ABCG2)+, C-X-C chemokine receptor type 4 (CXCR4)+ MSCs were differentiated after priming with ß-mercaptoethanol (ß-ME) combined with trans-retinoic acid (RA) and culture in neural basal media containing basic fibroblast growth factor (FGF2) and epidermal growth factor (EGF) or co-culture with neuronal cell lines. Differentiation efficiencies in vitro were determined using flow cytometry or fluorescent microscopy of cytospins made of FACS sorted positive cells after staining for markers of immature or mature neuronal lineages. RA-primed CD133+ABCG2+CXCR4+ human MSCs were transplanted into the lateral ventricle of male Sprague-Dawley rats, 24 hours after sham or traumatic brain injury (TBI). All animals were evaluated for spatial memory performance using the Morris Water Maze (MWM) Test. Histological examination of sham or TBI brains was done to evaluate MSC survival, migration and differentiation into neural lineages. We also examined induction of apoptosis at the injury site and production of MSC neuroprotective factors. RESULTS: CD133+ABCG2+CXCR4+ MSCs consistently expressed markers of neural lineage induction and were positive for nestin, microtubule associated protein-1ß (MAP-1ß), tyrosine hydroxylase (TH), neuron specific nuclear protein (NEUN) or type III beta-tubulin (Tuj1). Animals in the primed MSC treatment group exhibited MWM latency results similar to the uninjured (sham) group with both groups showing improvements in latency. Histological examination of brains of these animals showed that in uninjured animals the majority of MSCs were found in the lateral ventricle, the site of transplantation, while in TBI rats MSCs were consistently found in locations near the injury site. We found that levels of apoptosis were less in MSC treated rats and that MSCs could be shown to produce neurotropic factors as early as 2 days following transplantation of cells. In TBI rats, at 1 and 3 months post transplantation cells were generated which expressed markers of neural lineages including immature as well as mature neurons. CONCLUSIONS: These results suggest that PBD CD133+ABCG2+CXCR4+ MSCs have the potential for development as an autologous treatment for TBI and neurodegenerative disorders and that MSC derived cell products produced immediately after transplantation may aid in reducing the immediate cognitive defects of TBI.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antígenos CD/metabolismo , Apoptosis/fisiología , Lesiones Encefálicas/metabolismo , Glicoproteínas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fármacos Neuroprotectores/metabolismo , Péptidos/metabolismo , Receptores CXCR4/metabolismo , Antígeno AC133 , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Neurogénesis/fisiología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
14.
Stem Cell Res Ther ; 4 Suppl 1: S7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24565038

RESUMEN

Detailed studies of lung pathology in patients during the course of development of acute lung injury or respiratory distress are limited, and in the past information related to lung-specific responses has been derived from the study of lungs from patients who died at autopsy or from animal models. Development of good in vitro human tissue models would help to bridge the gap in our current knowledge of lung responses and provide a better understanding of lung development, physiology and pathology. In vitro models of simple one-cell or two-cell culture systems as well as complex multicellular lung analogs that reproduce defined components of specific human lung responses have already been realized. A benefit of current in vitro lung models is that hypotheses generated from review of data from human or animal disease studies can be tested directly in engineered human tissue models. Results of studies done using simple in vitro lung systems or more complex three-dimensional models have already been used to examine cell-based responses, physiologic functions, pathologic changes and even drug toxicity or drug responses. In the future we will create models with specific genetic profiles to test the importance of single gene products or pathways of significance. Recent development of microfluidics-based models that support high-throughput screening will allow early-stage toxicity testing in human systems and faster development of new and innovative medical products. Model design in the future will also allow for evaluation of multiple organ systems at once, providing a more holistic or whole-body approach to understanding human physiology and responses.


Asunto(s)
Enfermedades Pulmonares/patología , Pulmón/fisiología , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Enfermedades Pulmonares/metabolismo , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Modelos Biológicos , Proteína A Asociada a Surfactante Pulmonar/metabolismo
15.
J Cell Biochem ; 113(7): 2185-92, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22573544

RESUMEN

Pulmonary disease is a worldwide public health problem that reduces the quality of life and increases the need for hospital admissions as well as the risk for premature death for those affected. For many patients, lung transplantation is the only chance for survival. Unfortunately, there is a significant shortage of lungs for transplantation and since the lung is the most likely organ to be damaged during procurement many lungs deemed unacceptable for transplantation are simply discarded. Rather than discarding these lungs they can be used to produce three-dimensional acellular (AC) natural lung scaffolds for the generation of engineered lung tissue. AC scaffolds are lungs whose original cells have been destroyed by exposure to detergents and physical methods of removing cells and cell debris. This creates a lung scaffold from the skeleton of the lungs themselves. The scaffolds are then used to support adult, stem or progenitor cells which can be grown into functional lung tissue. Recent studies show that engineered lung tissues are capable of surviving after in vivo transplantation and support limited gas exchange. In the future engineered lung tissue has the potential to be used in clinical applications to replace lung functions lost following injury or disease. This manuscript discusses recent advances in development and use of AC scaffolds to support engineering of lung tissues.


Asunto(s)
Enfermedades Pulmonares/terapia , Pulmón/citología , Pulmón/cirugía , Ingeniería de Tejidos/métodos , Andamios del Tejido , Células Madre Adultas/metabolismo , Animales , Matriz Extracelular/fisiología , Humanos , Pulmón/fisiología , Enfermedades Pulmonares/patología , Trasplante de Pulmón/métodos , Ratas , Ratas Sprague-Dawley
16.
Tissue Eng Part A ; 16(8): 2565-80, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20408765

RESUMEN

We report here the first attempt to produce and use whole acellular (AC) lung as a matrix to support development of engineered lung tissue from murine embryonic stem cells (mESCs). We compared the influence of AC lung, Gelfoam, Matrigel, and a collagen I hydrogel matrix on the mESC attachment, differentiation, and subsequent formation of complex tissue. We found that AC lung allowed for better retention of cells with more differentiation of mESCs into epithelial and endothelial lineages. In constructs produced on whole AC lung, we saw indications of organization of differentiating ESC into three-dimensional structures reminiscent of complex tissues. We also saw expression of thyroid transcription factor-1, an immature lung epithelial cell marker; pro-surfactant protein C, a type II pneumocyte marker; PECAM-1/CD31, an endothelial cell marker; cytokeratin 18; alpha-actin, a smooth muscle marker; CD140a or platelet-derived growth factor receptor-alpha; and Clara cell protein 10. There was also evidence of site-specific differentiation in the trachea with the formation of sheets of cytokeratin-positive cells and Clara cell protein 10-expressing Clara cells. Our findings support the utility of AC lung as a matrix for engineering lung tissue and highlight the critical role played by matrix or scaffold-associated cues in guiding ESC differentiation toward lung-specific lineages.


Asunto(s)
Sistema Libre de Células/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Matriz Extracelular/metabolismo , Pulmón/fisiología , Técnicas de Cultivo de Órganos/métodos , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Células Cultivadas , Ratones , Ratas
17.
Regen Med ; 5(2): 289-98, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20210588

RESUMEN

Bone marrow is the primary site of hematopoiesis in adult humans. Bone marrow can be cultured in vitro but few simple culture systems fully support hematopoiesis beyond a few months. Human bone marrow analogs are long-term in vitro cultures of marrow stromal and hematopoietic stem cells that can be used to produce cells and products normally harvested from human donors. Bone marrow analog systems should exhibit confluence of the stromal cell populations, persistence of hematopoietic progenitor cells, presence of active regions of hematopoiesis and capacity to produce mature cell types for extended periods of time. Although we are still years away from realizing clinical application of products formed by artificial bone marrow analogs, the process of transitioning this research tool from bench to bedside should be fairly straightforward. The most obvious application of artificial marrow would be for production of autologous hematopoietic CD34(+) stem cells as a stem cell therapy for individuals experiencing bone marrow failure due to disease or injury. Another logical application is for 'blood farming', a process for large-scale in vitro production of red blood cells, white blood cells or platelets, for transfusion or treatment. Other possibilities include production of nonhematopoietic stem cells such as osteogenic stromal cells, osteoblasts and rare pluripotent stem cells. Bone marrow analogs also have great potential as ex vivo human test systems and could play a critical role in drug discovery, drug development and toxicity testing in the future.


Asunto(s)
Órganos Artificiales , Médula Ósea/fisiología , Ingeniería de Tejidos , Eritrocitos/citología , Humanos , Modelos Biológicos , Control Social Formal , Células Madre/citología
18.
Organogenesis ; 5(2): 57-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19794900

RESUMEN

Before we can realize our long term goal of engineering lung tissue worthy of clinical applications, advances in the identification and utilization of cell sources, development of standardized procedures for differentiation of cells, production of matrix tailored to meet the needs of the lung and design of methods or techniques of applying the engineered tissues into the injured lung environment will need to occur. Design of better biomaterials with the capacity to guide stem cell behavior and facilitate lung lineage choice as well as seamlessly integrate with living lung tissue will be achieved through advances in the development of decellularized matrices and new understandings related to the influence of extracellular matrix on cell behavior and function. We have strong hopes that recent developments in the engineering of conducting airway from decellularized trachea will lead to similar breakthroughs in the engineering of distal lung components in the future.

19.
Biomaterials ; 30(6): 1071-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19042018

RESUMEN

In vitro replicas of bone marrow can potentially provide a continuous source of blood cells for transplantation and serve as a laboratory model to examine human immune system dysfunctions and drug toxicology. Here we report the development of an in vitro artificial bone marrow based on a 3D scaffold with inverted colloidal crystal (ICC) geometry mimicking the structural topology of actual bone marrow matrix. To facilitate adhesion of cells, scaffolds were coated with a layer of transparent nanocomposite. After seeding with hematopoietic stem cells (HSCs), ICC scaffolds were capable of supporting expansion of CD34+ HSCs with B-lymphocyte differentiation. Three-dimensional organization was shown to be critical for production of B cells and antigen-specific antibodies. Functionality of bone marrow constructs was confirmed by implantation of matrices containing human CD34+ cells onto the backs of severe combined immunodeficiency (SCID) mice with subsequent generation of human immune cells.


Asunto(s)
Materiales Biomiméticos , Médula Ósea/anatomía & histología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Linfocitos B/citología , Diferenciación Celular , Línea Celular , Coloides , Cristalización , Humanos , Ratones , Ratones SCID , Microscopía Electrónica de Rastreo , Células Madre/citología
20.
Tissue Eng ; 12(5): 1213-25, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16771635

RESUMEN

In this study, we describe the isolation and characterization of a population of adult-derived or somatic lung progenitor cells (SLPC) from adult mammalian lung tissue and the promotion of alveolar tissue growth by these cells (both in vitro and in vivo) after seeding onto synthetic polymer scaffolds. After extended in vitro culture, differentiating cells expressed Clara cell 10kDa protein, surfactant protein-C, and cytokeratin but did not form organized structures. When cells were combined with synthetic scaffolds, polyglycolic acid (PGA) or Pluronic F-127 (PF-127), and maintained in vitro or implanted in vivo, they expressed lung-specific markers for Clara cells, pneumocytes, and respiratory epithelium and organized into identifiable pulmonary structures (including those similar to alveoli and terminal bronchi), with evidence of smooth muscle development. Although PGA has been shown to be an excellent polymer for culture of specific cell types in vitro, in vivo culture in an immunocompetent host induced a foreign body response that altered the integrity of the developing lung tissue. Use of PF-127/cell constructs resulted in the development of tissue with less inflammatory reaction. These data suggest that the therapeutic use of engineered tissues requires both the use of specific cell phenotypes, as well as the careful selection of synthetic polymers, to facilitate the assembly of functional tissue.


Asunto(s)
Materiales Biocompatibles , Hidrogeles , Pulmón , Poloxámero , Ácido Poliglicólico , Trasplante de Células Madre , Células Madre , Ingeniería de Tejidos , Animales , Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/fisiología , Femenino , Pulmón/crecimiento & desarrollo , Pulmón/ultraestructura , Ratones , Ratones Desnudos , Microscopía Electrónica de Rastreo , Ovinos , Células Madre/metabolismo , Células Madre/ultraestructura , Trasplante Heterólogo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...