Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Stem Cells ; 32(6): 1616-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24449042

RESUMEN

Adipose-derived stromal/stem cells (ASCs) have anti-inflammatory as well as immunosuppressive activities and are currently the focus of clinical trials for a number of inflammatory diseases. Acute lung injury (ALI) is an inflammatory condition of the lung for which standard treatment is mainly supportive due to lack of effective therapies. Our recent studies have demonstrated the ability of both human ASCs (hASCs) and mouse ASCs (mASCs) to attenuate lung damage and inflammation in a rodent model of lipopolysaccharide-induced ALI, suggesting that ASCs may also be beneficial in treating ALI. To better understand how ASCs may act in ALI and to elucidate the mechanism(s) involved in ASC modulation of lung inflammation, gene expression analysis was performed in ASC-treated (hASCs or mASCs) and control sham-treated lungs. The results revealed a dramatic difference between the expression of anti-inflammatory molecules by hASCs and mASCs. These data show that the beneficial effects of hASCs and mASCs in ALI may result from the production of different paracrine factors. Interleukin 6 (IL-6) expression in the mASC-treated lungs was significantly elevated as compared to sham-treated controls 20 hours after delivery of the cells by oropharyngeal aspiration. Knockdown of IL-6 expression in mASCs by RNA interference abrogated most of their therapeutic effects, suggesting that the anti-inflammatory properties of mASCs in ALI are explained, at least in part, by activation of IL-6 secretion.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/terapia , Tejido Adiposo/citología , Interleucina-6/metabolismo , Trasplante de Células Madre , Células Madre/citología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Albúminas/metabolismo , Animales , Antiinflamatorios/metabolismo , Líquido del Lavado Bronquioalveolar , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Factor Inhibidor de Leucemia/metabolismo , Lipopolisacáridos , Pulmón/patología , Ratones Endogámicos C57BL , Células del Estroma
2.
J Vasc Res ; 51(3): 163-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24853910

RESUMEN

Therapies aimed at manipulating the microcirculation require the ability to control angiogenesis, defined as the sprouting of new capillaries from existing vessels. Blocking angiogenesis would be beneficial in many pathologies (e.g. cancer, retinopathies and rheumatoid arthritis). In others (e.g. myocardial infarction, stroke and hypertension), promoting angiogenesis would be desirable. We know that vascular pericytes elongate around endothelial cells (ECs) and are functionally associated with regulating vessel stabilization, vessel diameter and EC proliferation. During angiogenesis, bidirectional pericyte-EC signaling is critical for capillary sprout formation. Observations of pericytes leading capillary sprouts also implicate their role in EC guidance. As such, pericytes have recently emerged as a therapeutic target to promote or inhibit angiogenesis. Advancing our basic understanding of pericytes and developing pericyte-related therapies are challenged, like in many other fields, by questions regarding cell identity. This review article discusses what we know about pericyte phenotypes and the opportunity to advance our understanding by defining the specific pericyte cell populations involved in capillary sprouting.


Asunto(s)
Neovascularización Patológica , Neovascularización Fisiológica/fisiología , Pericitos/fisiología , Animales , Antígenos/metabolismo , Biomarcadores/metabolismo , Capilares/fisiología , Movimiento Celular , Embrión de Pollo , Células Endoteliales , Humanos , Ratones , Microcirculación , Fenotipo , Proteoglicanos/metabolismo , Ratas , Transducción de Señal , Tubulina (Proteína)/metabolismo
3.
Int J Cancer ; 127(8): 1741-7, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20521250

RESUMEN

Human cathelicidin LL-37, a host defense peptide derived from leukocytes and epithelial cells, plays a crucial role in innate and adaptive immunity. Not only does LL-37 eliminate pathogenic microbes directly but also modulates host immune responses. Emerging evidence from tumor biology studies indicates that LL-37 plays a prominent and complex role in carcinogenesis. Although overexpression of LL-37 has been implicated in the development or progression of many human malignancies, including breast, ovarian and lung cancers, LL-37 suppresses tumorigenesis in gastric cancer. These data are beginning to unveil the intricate and contradictory functions of LL-37. The reasons for the tissue-specific function of LL-37 in carcinogenesis remain to be elucidated. Here, we review the relationship between LL-37, its fragments and cancer progression as well as discuss the potential therapeutic implications of targeting this peptide.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Neoplasias/metabolismo , Humanos , Lipopolisacáridos/fisiología , Lipopolisacáridos/uso terapéutico , Neoplasias/tratamiento farmacológico , Catelicidinas
4.
Brain Res ; 1726: 146517, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31634451

RESUMEN

We suggested previously that hippocampal slices were protected from hypoxic depolarization and swelling by preincubating them at room temperature (Kreisman et al., 2000). We postulated that hypothermic preconditioning induced tolerance in our slices, which protected against hypoxic depolarization and swelling. Control hippocampal slices were incubated at 34-35 °C for two hours and the response to 10 min of severe hypoxia was compared to slices which were preconditioned for two hours at room temperature (22-23 °C) prior to warming to 34-35 °C. Recordings of the extracellular DC potential provided an index of tissue depolarization and changes in tissue light transmittance provided an index of swelling. Hypothermic preconditioning significantly reduced hypoxia-induced swelling, particularly in CA3 and the dentate inner blade. Since erythropoietin (EPO) had been shown to mediate hypoxic preconditioning, we tested whether EPO also mediated hypothermic preconditioning in our slices. Recombinant rat EPO (1-10 micromolar) mitigated hypoxia-induced swelling and depolarization in dentate inner blade of unconditioned slices in a dose-dependent manner. We also blocked the protective effects of hypothermic preconditioning on hypoxic depolarization and swelling in the inner blade of the dentate gyrus by administering soluble EPO receptor in the bath and treating slices with wortmannin to block phosphorylation of PI3 kinase, a critical step in the activation of the downstream neuroprotectant, Akt. These results suggest that EPO mediates tolerance to hypoxic depolarization and swelling induced by hypothermic preconditioning. They also emphasize that various preincubation protocols used in experiments with hippocampal slices may differentially affect basal electrophysiological and metabolic properties of those slices.


Asunto(s)
Eritropoyetina/administración & dosificación , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Hipotermia/fisiopatología , Hipoxia/fisiopatología , Precondicionamiento Isquémico , Fármacos Neuroprotectores/administración & dosificación , Animales , Hipocampo/patología , Hipotermia/patología , Hipoxia/patología , Masculino , Ratas Sprague-Dawley
5.
Tissue Eng Part A ; 20(9-10): 1426-43, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24378017

RESUMEN

There are an insufficient number of donor organs available to meet the demand for lung transplantation. This issue could be addressed by regenerating functional tissue from diseased or damaged lungs that would otherwise be deemed unsuitable for transplant. Detergent-mediated whole-lung decellularization produces a three-dimensional natural scaffold that can be repopulated with various cell types. In this study, we investigated the decellularization and initial recellularization of diseased lungs using a rat model of monocrotaline-induced pulmonary hypertension (MCT-PHT). Decellularization of control and MCT-PHT Sprague-Dawley rat lungs was accomplished by treating the lungs with a combination of Triton X-100, sodium deoxycholate, NaCl, and DNase. The resulting acellular matrices were characterized by DNA quantification, Western blotting, immunohistochemistry, and proteomic analyses revealing that decellularization was able to remove cells while leaving the extracellular matrix (ECM) components and lung ultrastructure intact. Decellularization significantly reduced DNA content (∼30-fold in MCT-PHT lungs and ∼50-fold in the control lungs) and enriched ECM components (>60-fold in both the control and MCT-PHT lungs) while depleting cellular proteins. MicroCT visualization of MCT-PHT rat lungs indicated that the vasculature was narrowed as a result of MCT treatment, and this characteristic was unchanged by decellularization. Mean arterial vessel diameter of representative decellularized MCT-PHT and control scaffolds was estimated to be 0.152±0.134 mm and 0.247±0.160 mm, respectively. Decellularized MCT-PHT lung scaffolds supported attachment and survival of rat adipose-derived stem cells (rASCs), seeded into the airspace or the vasculature, for at least 2 weeks. The cells seeded in MCT-PHT lung scaffolds proliferated and underwent apoptosis similar to control scaffolds; however, the initial percentage of apoptotic cells was slightly higher in MCT-PHT lungs (2.79±2.03% vs. 1.05±1.02% of airway-seeded rASCs, and 4.47±1.21% vs. 2.66±0.10% of vascular seeded rASCs). The ECM of cell-seeded scaffolds showed no signs of degradation by the cells after 14 days in culture. These data suggest that diseased hypertensive lungs can be efficiently decellularized similar to control lungs and have the potential to be recellularized with mesenchymal stem cells with the ultimate goal of generating healthy, functional pulmonary tissue.


Asunto(s)
Sistema Libre de Células/química , Hipertensión Pulmonar/patología , Pulmón/química , Pulmón/crecimiento & desarrollo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Andamios del Tejido , Animales , Órganos Bioartificiales , Proliferación Celular , Células Cultivadas , Diseño de Equipo , Matriz Extracelular/química , Hipertensión Pulmonar/metabolismo , Masculino , Técnicas de Cultivo de Órganos/métodos , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos/instrumentación
6.
Adv Biochem Eng Biotechnol ; 130: 163-97, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22869086

RESUMEN

Cell-based therapies (CBTs) are quickly taking hold as a revolutionary new approach to treat many human diseases. Among the cells used in these treatments, multipotent mesenchymal stromal cells, also often and imprecisely termed mesenchymal stem cells (MSC), are widely used because they are considered clinically safe, unique in their immune-modulating capabilities, easily obtained from adult tissues, and quickly expanded as well as stored. However, despite these established advantages, there are limiting factors to employing MSCs in these therapeutic strategies. Foremost is the lack of a general consensus on a definition of these cells, marring efforts to prepare homogeneous lots and more importantly complicating their in vitro and in vivo investigation. Furthermore, although one of the most profound clinical effects of MSC intravenous administration is the modulation of host immune responses, no adequate ex vivo assays exist to consistently predict the therapeutic effect of each MSC lot in the treated patient. Until these issues are addressed, this very promising and safe new therapeutic approach cannot be used to its full advantage. However, these confounding issues do present exciting opportunities. The first is an opportunity to discover unknown aspects of host immune responses because the unique effect driven by MSC infusion on a patient's immunity has not yet been identified. In addition, there is an opportunity to develop methods, tests, and tools to better define MSCs and MSC-based therapy and provide consistency in preparation and effect. To this end, my laboratory recently developed a new approach to induce uniform pro-inflammatory MSC1 and anti-inflammatory MSC2 phenotypes from bone marrow-derived MSC preparations. I anticipate that MSC1 and MSC2 provide convenient tools with which to address some of these limitations and will help advance safe and effective CBTs for human disease.

7.
J Vis Exp ; (82): e50825, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24378384

RESUMEN

There are an insufficient number of lungs available to meet current and future organ transplantation needs. Bioartificial tissue regeneration is an attractive alternative to classic organ transplantation. This technology utilizes an organ's natural biological extracellular matrix (ECM) as a scaffold onto which autologous or stem/progenitor cells may be seeded and cultured in such a way that facilitates regeneration of the original tissue. The natural ECM is isolated by a process called decellularization. Decellularization is accomplished by treating tissues with a series of detergents, salts, and enzymes to achieve effective removal of cellular material while leaving the ECM intact. Studies conducted utilizing decellularization and subsequent recellularization of rodent lungs demonstrated marginal success in generating pulmonary-like tissue which is capable of gas exchange in vivo. While offering essential proof-of-concept, rodent models are not directly translatable to human use. Nonhuman primates (NHP) offer a more suitable model in which to investigate the use of bioartificial organ production for eventual clinical use. The protocols for achieving complete decellularization of lungs acquired from the NHP rhesus macaque are presented. The resulting acellular lungs can be seeded with a variety of cells including mesenchymal stem cells and endothelial cells. The manuscript also describes the development of a bioreactor system in which cell-seeded macaque lungs can be cultured under conditions of mechanical stretch and strain provided by negative pressure ventilation as well as pulsatile perfusion through the vasculature; these forces are known to direct differentiation along pulmonary and endothelial lineages, respectively. Representative results of decellularization and cell seeding are provided.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/trasplante , Reactores Biológicos , Células Endoteliales/citología , Pulmón/fisiología , Regeneración/fisiología , Ingeniería de Tejidos/métodos , Animales , Separación Celular/métodos , Pulmón/citología , Macaca mulatta , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/instrumentación
8.
Stem Cell Res Ther ; 4(1): 13, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23360775

RESUMEN

INTRODUCTION: Adipose-derived stem cells (ASCs) have emerged as important regulators of inflammatory/immune responses in vitro and in vivo and represent attractive candidates for cell-based therapies for diseases that involve excessive inflammation. Acute lung injury (ALI) is an inflammatory condition for which treatment is mainly supportive due to lack of effective therapies. In this study, the therapeutic effects of ASC-based therapy were assessed in vivo by comparison of the anti-inflammatory properties of both human and murine ASCs in a mouse model of lipopolysaccharide (LPS)-induced ALI. METHODS: Human ASCs (hASCs) or mouse ASCs (mASCs) were delivered to C57Bl/6 mice (7.5 × 105 total cells/mouse) by oropharyngeal aspiration (OA) four hours after the animals were challenged with lipopolysaccharide (15 mg/kg). Mice were sacrificed 24 and 72 hours after LPS exposure, and lung histology examined for evaluation of inflammation and injury. Bronchoalveolar lavage fluid (BALF) was analyzed to determine total and differential cell counts, total protein and albumin concentrations, and myeloperoxidase (MPO) activity. Cytokine expression in the injured lungs was measured at the steady-state mRNA levels and protein levels for assessment of the degree of lung inflammation. RESULTS: Both human and mouse ASC treatments provided protective anti-inflammatory responses. There were decreased levels of leukocyte (for example neutrophil) migration into the alveoli, total protein and albumin concentrations in BALF, and MPO activity after the induction of ALI following both therapies. Additionally, cell therapy with both cell types effectively suppressed the expression of proinflammatory cytokines and increased the anti-inflammatory cytokine interleukin 10 (IL-10). Overall, the syngeneic mASC therapy had a more potent therapeutic effect than the xenogeneic hASC therapy in this model. CONCLUSIONS: Treatment with hASCs or mASCs significantly attenuated LPS-induced acute lung injury in mice. These results suggest a potential benefit for using an ASC-based therapy to treat clinical ALI and may possibly prevent the development of acute respiratory distress syndrome (ARDS).


Asunto(s)
Lesión Pulmonar Aguda/fisiopatología , Tejido Adiposo/fisiología , Lipopolisacáridos/farmacología , Células Madre/fisiología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Tejido Adiposo/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-10/metabolismo , Leucocitos/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Peroxidasa/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/fisiopatología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/fisiopatología , Células Madre/metabolismo
9.
PLoS One ; 7(9): e45590, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029122

RESUMEN

BACKGROUND: Currently, there are many promising clinical trials using mesenchymal stem cells (MSCs) in cell-based therapies of numerous diseases. Increasingly, however, there is a concern over the use of MSCs because they home to tumors and can support tumor growth and metastasis. For instance, we established that MSCs in the ovarian tumor microenvironment promoted tumor growth and favored angiogenesis. In parallel studies, we also developed a new approach to induce the conventional mixed pool of MSCs into two uniform but distinct phenotypes we termed MSC1 and MSC2. METHODOLOGY/PRINCIPAL FINDINGS: Here we tested the in vitro and in vivo stability of MSC1 and MSC2 phenotypes as well as their effects on tumor growth and spread. In vitro co-culture of MSC1 with various cancer cells diminished growth in colony forming units and tumor spheroid assays, while conventional MSCs or MSC2 co-culture had the opposite effect in these assays. Co-culture of MSC1 and cancer cells also distinctly affected their migration and invasion potential when compared to MSCs or MSC2 treated samples. The expression of bioactive molecules also differed dramatically among these samples. MSC1-based treatment of established tumors in an immune competent model attenuated tumor growth and metastasis in contrast to MSCs- and MSC2-treated animals in which tumor growth and spread was increased. Also, in contrast to these groups, MSC1-therapy led to less ascites accumulation, increased CD45+leukocytes, decreased collagen deposition, and mast cell degranulation. CONCLUSION/SIGNIFICANCE: These observations indicate that the MSC1 and MSC2 phenotypes may be convenient tools for the discovery of critical components of the tumor stroma. The continued investigation of these cells may help ensure that cell based-therapy is used safely and effectively in human disease.


Asunto(s)
División Celular , Trasplante de Células Madre Mesenquimatosas , Metástasis de la Neoplasia , Neoplasias/cirugía , Línea Celular Tumoral , Técnicas de Cocultivo , Humanos , Neoplasias/patología
10.
PLoS One ; 7(6): e39592, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22745793

RESUMEN

BACKGROUND: The use of bone marrow-derived human multipotent stromal cells (hMSC) in cell-based therapies has dramatically increased in recent years, as researchers have exploited the ability of these cells to migrate to sites of tissue injury, inflammation, and tumors. Our group established that hMSC respond to "danger" signals--by-products of damaged, infected or inflamed tissues--via activation of Toll-like receptors (TLRs). However, little is known regarding downstream signaling mediated by TLRs in hMSC. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate that TLR3 stimulation activates a Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 1 pathway, and increases expression of suppressor of cytokine signaling (SOCS) 1 and SOCS3 in hMSC. Our studies suggest that each of these SOCS plays a distinct role in negatively regulating TLR3 and JAK/STAT signaling. TLR3-mediated interferon regulatory factor 1 (IRF1) expression was inhibited by SOCS3 overexpression in hMSC while SOCS1 overexpression reduced STAT1 activation. Furthermore, our study is the first to demonstrate that when TLR3 is activated in hMSC, expression of CXCR4 and CXCR7 is downregulated. SOCS3 overexpression inhibited internalization of both CXCR4 and CXCR7 following TLR3 stimulation. In contrast, SOCS1 overexpression only inhibited CXCR7 internalization. CONCLUSION/SIGNIFICANCE: These results demonstrate that SOCS1 and SOCS3 each play a functionally distinct role in modulating TLR3, JAK/STAT, and CXCR4/CXCR7 signaling in hMSC and shed further light on the way hMSC respond to danger signals.


Asunto(s)
Células de la Médula Ósea/citología , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 3/metabolismo , Células Cultivadas , Humanos , Receptores CXCR/genética , Receptores CXCR4/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Receptor Toll-Like 3/genética
11.
J Vis Exp ; (63): e3954, 2012 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-22643964

RESUMEN

Microvacular network growth and remodeling are critical aspects of wound healing, inflammation, diabetic retinopathy, tumor growth and other disease conditions. Network growth is commonly attributed to angiogenesis, defined as the growth of new vessels from pre-existing vessels. The angiogenic process is also directly linked to arteriogenesis, defined as the capillary acquisition of a perivascular cell coating and vessel enlargement. Needless to say, angiogenesis is complex and involves multiple players at the cellular and molecular level. Understanding how a microvascular network grows requires identifying the spatial and temporal dynamics along the hierarchy of a network over the time course of angiogenesis. This information is critical for the development of therapies aimed at manipulating vessel growth. The exteriorization model described in this article represents a simple, reproducible model for stimulating angiogenesis in the rat mesentery. It was adapted from wound-healing models in the rat mesentery, and is an alternative to stimulate angiogenesis in the mesentery via i.p. injections of pro-angiogenic agents. The exteriorization model is attractive because it requires minimal surgical intervention and produces dramatic, reproducible increases in capillary sprouts, vascular area and vascular density over a relatively short time course in a tissue that allows for the two-dimensional visualization of entire microvascular networks down to single cell level. The stimulated growth reflects natural angiogenic responses in a physiological environment without interference of foreign angiogenic molecules. Using immunohistochemical labeling methods, this model has been proven extremely useful in identifying novel cellular events involved in angiogenesis. Investigators can readily correlate the angiogenic metrics during the time course of remodeling with time specific dynamics, such as cellular phenotypic changes or cellular interactions.


Asunto(s)
Mesenterio/irrigación sanguínea , Mesenterio/cirugía , Neovascularización Fisiológica/fisiología , Animales , Masculino , Ratas , Ratas Wistar
12.
Stem Cells Transl Med ; 1(7): 557-65, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23197860

RESUMEN

Mesenchymal stem cells (MSCs) are very attractive candidates in cell-based strategies that target inflammatory diseases. Preclinical animal studies and many clinical trials have demonstrated that human MSCs can be safely administered and that they modify the inflammatory process in the targeted injured tissue. Our laboratory developed a novel method that optimizes the anti-inflammatory effects of MSCs. We termed the cells prepared by this method MSC2. In this study, we determined the effects of MSC2-based therapies on an inflammation-linked painful diabetic peripheral neuropathy (pDPN) mouse model. Streptozotocin-induced diabetic mice were treated with conventionally prepared MSCs, MSC2, or vehicle at three specific time points. Prior to each treatment, responses to radiant heat (Hargreaves) and mechanical stimuli (von Frey) were measured. Blood serum from each animal was collected at the end of the study to compare levels of inflammatory markers between the treatment groups. We observed that MSC2-treated mice had significant improvement in behavioral assays compared with the vehicle and MSC groups, and moreover these responses did not differ from the observations seen in the healthy wild-type control group. Mice treated with conventional MSCs showed significant improvement in the radiant heat assay, but not in the von Frey test. Additionally, mice treated with MSC2 had decreased serum levels in many proinflammatory cytokines compared with the values measured in the MSC- or vehicle-treated groups. These findings indicate that MSC2-based therapy is a new anti-inflammatory treatment to consider in the management of pDPN.


Asunto(s)
Citocinas/sangre , Diabetes Mellitus Experimental/terapia , Neuropatías Diabéticas/terapia , Mediadores de Inflamación/sangre , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Animales , Biomarcadores/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/patología , Neuropatías Diabéticas/sangre , Neuropatías Diabéticas/patología , Humanos , Inflamación/sangre , Inflamación/patología , Inflamación/terapia , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Trasplante Homólogo
13.
Tissue Eng Part A ; 18(23-24): 2437-52, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22764775

RESUMEN

Currently, patients with end-stage lung disease are limited to lung transplantation as their only treatment option. Unfortunately, the lungs available for transplantation are few. Moreover, transplant recipients require life-long immune suppression to tolerate the transplanted lung. A promising alternative therapeutic strategy is decellularization of whole lungs, which permits the isolation of an intact scaffold comprised of innate extracellular matrix (ECM) that can theoretically be recellularized with autologous stem or progenitor cells to yield a functional lung. Nonhuman primates (NHP) provide a highly relevant preclinical model with which to assess the feasibility of recellularized lung scaffolds for human lung transplantation. Our laboratory has successfully accomplished lung decellularization and initial stem cell inoculation of the resulting ECM scaffold in an NHP model. Decellularization of normal adult rhesus macaque lungs as well as the biology of the resulting acellular matrix have been extensively characterized. Acellular NHP matrices retained the anatomical and ultrastructural properties of native lungs with minimal effect on the content, organization, and appearance of ECM components, including collagen types I and IV, laminin, fibronectin, and sulfated glycosaminoglycans (GAG), due to decellularization. Proteomics analysis showed enrichment of ECM proteins in total tissue extracts due to the removal of cells and cellular proteins by decellularization. Cellular DNA was effectively removed after decellularization (∼92% reduction), and the remaining nuclear material was found to be highly disorganized, very-low-molecular-weight fragments. Both bone marrow- and adipose-derived mesenchymal stem cells (MSC) attach to the decellularized lung matrix and can be maintained within this environment in vitro, suggesting that these cells may be promising candidates and useful tools for lung regeneration. Analysis of decellularized lung slice cultures to which MSC were seeded showed that the cells attached to the decellularized matrix, elongated, and proliferated in culture. Future investigations will focus on optimizing the recellularization of NHP lung scaffolds toward the goal of regenerating pulmonary tissue. Bringing this technology to eventual human clinical application will provide patients with an alternative therapeutic strategy as well as significantly reduce the demand for transplantable organs and patient wait-list time.


Asunto(s)
Pulmón/fisiología , Macaca mulatta/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Modelos Animales , Regeneración , Manejo de Especímenes/métodos , Andamios del Tejido , Animales , Apoptosis , Adhesión Celular , ADN/aislamiento & purificación , Ácido Desoxicólico/farmacología , Desoxirribonucleasas/farmacología , Detergentes/farmacología , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Proteínas de la Matriz Extracelular/análisis , Femenino , Fijadores/farmacología , Glicosaminoglicanos/análisis , Pulmón/química , Pulmón/efectos de los fármacos , Pulmón/ultraestructura , Macaca mulatta/anatomía & histología , Masculino , Perfusión , Proteómica , Solución Salina Hipertónica/farmacología , Andamios del Tejido/química
14.
Influenza Other Respir Viruses ; 5(Suppl 1): 198-201, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-29415086

RESUMEN

BACKGROUND: Increased susceptibility of older populations to secondary bacterial pneumonia-like infections following influenza infection has been well documented.1 Recent evidence in mouse models suggests that this increased risk from secondary bacterial infection occurs through a desensitization of the innate immune response.2 This recent finding, however, does not account for potential differences in immune responsiveness due to age. MATERIALS AND METHODS: To address this parameter, we used three age groups (aged, adult, and young mice) to evaluate the role of age in influenza-mediated vulnerability to secondary bacterial challenge with Pseudomonas aeruginosa. All mice were evaluated for multiple parameters including: (i) survival; (ii) lung bacterial load; (iii) total lung protein content; (iv) immune cell infiltration; (v) cytokine/chemokine expression; and (vi) toll-like receptor (TLR) RNA expression profiles. RESULTS: Prior challenge with influenza contributed to aberrant cytokine/chemokine profiles and increased lung cellular infiltrate in response to secondary bacterial infection across all age groups, supporting a critical role for influenza infection in the alteration of immune responses to other pathogens. Also similar to human influenza, these changes were exacerbated by age in mice as demonstrated by increased bacterial load, mortality, and total lung protein content (an indicator of lung damage) after P. aeruginosa challenge. CONCLUSIONS: These data support a potential role for virus-mediated and age-mediated alteration of innate immune effectors in the pathogenesis of influenza and the increased susceptibility of influenza virus infected mice to secondary bacterial infection. The understanding of the complex interaction of host and pathogen - and the role of age - in human influenza is critical in the development of novel therapeutics and improved vaccine approaches for influenza. Our results support further examination of influenza-mediated alterations in innate immune responses in aged and non-aged animals to allow elucidation of the molecular mechanisms of influenza pathogenesis in humans.

15.
Stem Cell Res Ther ; 1(5): 34, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21092149

RESUMEN

Mesenchymal stem cells (MSCs) are the nonhematopoietic multipotent progenitor cells found in various adult tissues. They are characterized by their ease of isolation and their rapid growth in vitro while maintaining their differentiation potential, allowing for extensive expansion in culture that yields large quantities suitable for therapeutic use. This article reviews the immunomodulatory activities associated with MSCs. Numerous studies have demonstrated that MSCs are potently immunosuppressive in vitro and in vivo. However, this article presents a new paradigm in MSC biology, in which MSCs, at least in vitro, can undergo polarization into either a pro-inflammatory or an immunosuppressive phenotype.


Asunto(s)
Tolerancia Inmunológica/inmunología , Inmunomodulación/inmunología , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Células Madre Mesenquimatosas/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/citología
16.
PLoS One ; 5(4): e10088, 2010 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-20436665

RESUMEN

BACKGROUND: Our laboratory and others reported that the stimulation of specific Toll-like receptors (TLRs) affects the immune modulating responses of human multipotent mesenchymal stromal cells (hMSCs). Toll-like receptors recognize "danger" signals, and their activation leads to profound cellular and systemic responses that mobilize innate and adaptive host immune cells. The danger signals that trigger TLRs are released following most tissue pathologies. Since danger signals recruit immune cells to sites of injury, we reasoned that hMSCs might be recruited in a similar way. Indeed, we found that hMSCs express several TLRs (e.g., TLR3 and TLR4), and that their migration, invasion, and secretion of immune modulating factors is drastically affected by specific TLR-agonist engagement. In particular, we noted diverse consequences on the hMSCs following stimulation of TLR3 when compared to TLR4 by our low-level, short-term TLR-priming protocol. PRINCIPAL FINDINGS: Here we extend our studies on the effect on immune modulation by specific TLR-priming of hMSCs, and based on our findings, propose a new paradigm for hMSCs that takes its cue from the monocyte literature. Specifically, that hMSCs can be polarized by downstream TLR signaling into two homogenously acting phenotypes we classify here as MSC1 and MSC2. This concept came from our observations that TLR4-primed hMSCs, or MSC1, mostly elaborate pro-inflammatory mediators, while TLR3-primed hMSCs, or MSC2, express mostly immunosuppressive ones. Additionally, allogeneic co-cultures of TLR-primed MSCs with peripheral blood mononuclear cells (PBMCs) predictably lead to suppressed T-lymphocyte activation following MSC2 co-culture, and permissive T-lymphocyte activation in co-culture with MSC1. SIGNIFICANCE: Our study provides an explanation to some of the conflicting reports on the net effect of TLR stimulation and its downstream consequences on the immune modulating properties of stem cells. We further suggest that MSC polarization provides a convenient way to render these heterogeneous preparations of cells more uniform while introducing a new facet to study, as well as provides an important aspect to consider for the improvement of current stem cell-based therapies.


Asunto(s)
Inmunidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Movimiento Celular/inmunología , Técnicas de Cocultivo , Humanos , Sistema Inmunológico/citología , Inflamación/inmunología , Monocitos , Fenotipo , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/inmunología , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA