Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 10: 1228, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31214185

RESUMO

Rapid progress is occurring in understanding the mechanisms underlying mesenchymal stromal cell (MSC)-based cell therapies (MSCT). However, the results of clinical trials, while demonstrating safety, have been varied in regard to efficacy. Recent data from different groups have shown profound and significant influences of the host inflammatory environment on MSCs delivered systemically or through organ-specific routes, for example intratracheal, with subsequent actions on potential MSC efficacies. Intriguingly in some models, it appears that dead or dying cells or subcellular particles derived from them, may contribute to therapeutic efficacy, at least in some circumstances. Thus, the broad cellular changes that accompany MSC death, autophagy, pre-apoptotic function, or indeed the host response to these processes may be essential to therapeutic efficacy. In this review, we summarize the existing literature concerning the necrobiology of MSCs and the available evidence that MSCs undergo autophagy, apoptosis, transfer mitochondria, or release subcellular particles with effector function in pathologic or inflammatory in vivo environments. Advances in understanding the role of immune effector cells in cell therapy, especially macrophages, suggest that the reprogramming of immunity associated with MSCT has a weighty influence on therapeutic efficacy. If correct, these data suggest novel approaches to enhancing the beneficial actions of MSCs that will vary with the inflammatory nature of different disease targets and may influence the choice between autologous or allogeneic or even xenogeneic cells as therapeutics.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Apoptose , Autofagia , Transporte Biológico , Comunicação Celular/imunologia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Vesículas Extracelulares/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Mitocôndrias/metabolismo , Resultado do Tratamento
2.
Sci Rep ; 6: 38207, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27922052

RESUMO

Bone-marrow derived mesenchymal stromal cells (MSCs) have potent immunomodulatory and tissue reparative properties, which may be beneficial in the treatment of inflammatory diseases such as COPD. This study examined the mechanisms by which human MSCs protect against elastase induced emphysema. Using a novel human relevant pre-clinical model of emphysema the efficacy of human MSC therapy and optimal cell dose were investigated. Protective effects were examined in the lung through histological examination. Further in vivo experiments examined the reparative abilities of MSCs after tissue damage was established and the role played by soluble factors secreted by MSCs. The mechanism of MSC action was determined in using shRNA gene knockdown. Human MSC therapy and MSC conditioned media exerted significant cytoprotective effects when administered early at the onset of the disease. These protective effects were due to significant anti-inflammatory, anti-fibrotic and anti-apoptotic mechanisms, mediated in part through MSC production of hepatocyte growth factor (HGF). When MSC administration was delayed, significant protection of the lung architecture was observed but this was less extensive. MSC cell therapy was more effective than MSC conditioned medium in this emphysema model.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Doença Pulmonar Obstrutiva Crônica/prevenção & controle , Animais , Apoptose , Modelos Animais de Doenças , Enfisema/etiologia , Enfisema/terapia , Fibrose , Fator de Crescimento de Hepatócito/genética , Humanos , Pulmão/patologia , Camundongos Endogâmicos NOD , Elastase Pancreática/toxicidade , Doença Pulmonar Obstrutiva Crônica/etiologia
3.
Stem Cells Transl Med ; 5(10): 1307-1318, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27388243

RESUMO

: The incidence of idiopathic pulmonary fibrosis is on the rise and existing treatments have failed to halt or reverse disease progression. Mesenchymal stromal cells (MSCs) have potent cytoprotective effects, can promote tissue repair, and have demonstrated efficacy in a range of fibrotic lung diseases; however, the exact mechanisms of action remain to be elucidated. Chemical antagonists and short hairpin RNA knockdown were used to identify the mechanisms of action used by MSCs in promoting wound healing, proliferation, and inhibiting apoptosis. Using the bleomycin induced fibrosis model, the protective effects of early or late MSC administration were examined. The role for hepatocyte growth factor (HGF) in MSC protection against bleomycin lung injury was examined using HGF knockdown MSC. Terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling assay was performed on ex vivo lung sections to examine the effects of MSC on apoptosis. MSC conditioned media (CM) enhanced wound closure and inhibited apoptosis of pulmonary cells in vitro. HGF was required for MSC CM enhancement of epithelial cell proliferation and inhibition of apoptosis. In contrast, MSC required COX-2 for CM to inhibit fibroblast proliferation. In a murine model, early administration of MSC protected against bleomycin induced lung fibrosis and correlated with reduced levels of the proinflammatory cytokine interleukin-1ß, reduced levels of apoptosis, and significantly increased levels of HGF. These protective effects were in part mediated by MSC derived HGF as HGF knockdown MSC were unable to protect against fibrosis in vivo. These findings delineate the mechanisms of MSC protection in a preclinical model of fibrotic lung disease. SIGNIFICANCE: The mechanisms used by mesenchymal stromal cells (MSCs) in mediating protective effects in chronic models of lung disease are not understood and remain to be elucidated. These findings from in vitro studies highlight an important role for the MSC-derived soluble factors hepatocyte growth factor (HGF) and prostaglandin E2 in promoting wound healing and inhibiting apoptosis. Furthermore, this study translates these findings demonstrating an important role for HGF in the protective effects mediated by MSC in vivo in the bleomycin model. These findings support a targeted approach to enhancing MSC therapy for fibrotic disease and highlight the importance of timing of MSC therapy.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Fibrose Pulmonar Idiopática/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose/fisiologia , Bleomicina/toxicidade , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Fibrose Pulmonar Idiopática/metabolismo , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
4.
Infect Immun ; 84(5): 1424-1437, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902727

RESUMO

Members of the Burkholderia cepacia complex (Bcc) cause chronic opportunistic lung infections in people with cystic fibrosis (CF), resulting in a gradual lung function decline and, ultimately, patient death. The Bcc is a complex of 20 species and is rarely eradicated once a patient is colonized; therefore, vaccination may represent a better therapeutic option. We developed a new proteomics approach to identify bacterial proteins that are involved in the attachment of Bcc bacteria to lung epithelial cells. Fourteen proteins were reproducibly identified by two-dimensional gel electrophoresis from four Bcc strains representative of two Bcc species: Burkholderia cenocepacia, the most virulent, and B. multivorans, the most frequently acquired. Seven proteins were identified in both species, but only two were common to all four strains, linocin and OmpW. Both proteins were selected based on previously reported data on these proteins in other species. Escherichia coli strains expressing recombinant linocin and OmpW showed enhanced attachment (4.2- and 3.9-fold) to lung cells compared to the control, confirming that both proteins are involved in host cell attachment. Immunoproteomic analysis using serum from Bcc-colonized CF patients confirmed that both proteins elicit potent humoral responses in vivo Mice immunized with either recombinant linocin or OmpW were protected from B. cenocepacia and B. multivorans challenge. Both antigens induced potent antigen-specific antibody responses and stimulated strong cytokine responses. In conclusion, our approach identified adhesins that induced excellent protection against two Bcc species and are promising vaccine candidates for a multisubunit vaccine. Furthermore, this study highlights the potential of our proteomics approach to identify potent antigens against other difficult pathogens.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Bacteriocinas/metabolismo , Infecções por Burkholderia/prevenção & controle , Complexo Burkholderia cepacia/fisiologia , Células Epiteliais/microbiologia , Adesinas Bacterianas/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Bacteriocinas/imunologia , Infecções por Burkholderia/imunologia , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/fisiologia , Feminino , Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resultado do Tratamento
5.
Stem Cells Dev ; 24(20): 2391-402, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26076727

RESUMO

The immune suppressive and anti-inflammatory capabilities of bone marrow-derived mesenchymal stromal cells (MSCs) represent an innovative new tool in regenerative medicine and immune regulation. The potent immune suppressive ability of MSC over T cells, dendritic cells, and natural killer cells has been extensively characterized, however, the effect of MSC on B cell function has not yet been clarified. In this study, the direct effect of MSC on peripheral blood B cell function is defined and the mechanism utilized by MSC in enhancing B cell survival in vitro identified. Human MSC supported the activation, proliferation, and survival of purified CD19(+) B cells through a cell contact-dependent mechanism. These effects were not mediated through B cell activating factor or notch signaling. However, cell contact between MSC and B cells resulted in increased production of vascular endothelial growth factor (VEGF) by MSC facilitating AKT phosphorylation within the B cell and inhibiting caspase 3-mediated apoptosis. Blocking studies demonstrated that this cell contact-dependent effect was not dependent on signaling through CXCR4-CXCL12 or through the epidermal growth factor receptor (EGFR). These results suggest that direct cell contact between MSC and B cells supports B cell viability and function, suggesting that MSC may not represent a suitable therapy for B cell-mediated disease.


Assuntos
Apoptose/imunologia , Linfócitos B/citologia , Caspase 3/metabolismo , Células-Tronco Mesenquimais/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antígenos CD19/imunologia , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Proliferação de Células/fisiologia , Humanos , Células Matadoras Naturais/imunologia , Transplante de Células-Tronco Mesenquimais , Linfócitos T/citologia , Ativação Transcricional/imunologia , Regulação para Cima
6.
Stem Cell Res Ther ; 6: 19, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25890330

RESUMO

INTRODUCTION: Mesenchymal stromal cells (MSC) have well defined immunomodulatory properties including the suppression of lymphocyte proliferation and inhibition of dendritic cell (DC) maturation involving both cell contact and soluble factors. These properties have made MSC attractive candidates for cellular therapy. However, the mechanism underlying these characteristics remains unclear. This study sought to investigate the mechanisms by which MSC induce a regulatory environment. METHOD: Allogeneic bone marrow mesenchymal stromal cells were cultured with T cells or dendritic cells in the presence or absence of gamma secretase inhibitor to block Notch receptor signalling. T cells and dendritic cells were examined by flow cytometry for changes in phenotype marker expression. Stable knock down MSC were generated to examine the influence of Jagged 1 signalling by MSC. Both wildtype and knockdown MSC were subsequently used in vivo in an animal model of allergic airway inflammation. RESULTS: The Notch ligand Jagged-1 was demonstrated to be involved in MSC expansion of regulatory T cells (Treg). Additionally, MSC-induced a functional semi-mature DC phenotype, which further required Notch signalling for the expansion of Treg. MSC, but not Jagged-1 knock down MSC, reduced pathology in a mouse model of allergic airway inflammation. Protection mediated by MSC was associated with enhanced Treg in the lung and significantly increased production of interleukin (IL)-10 in splenocytes re-stimulated with allergen. Significantly less Treg and IL-10 was observed in mice treated with Jagged-1 knock down MSC. CONCLUSIONS: The current study suggests that MSC-mediated immune modulation involves the education and expansion of regulatory immune cells in a Jagged-1 dependent manner and provides the first report of the importance of Jagged-1 signalling in MSC protection against inflammation in vivo.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Células Dendríticas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/imunologia , Pneumonia/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular , Proliferação de Células , Células Dendríticas/citologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Tolerância Imunológica/imunologia , Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Proteína Jagged-1 , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Interferência de RNA , RNA Interferente Pequeno , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais , Linfócitos T Reguladores/citologia
7.
Curr Drug Deliv ; 11(5): 561-71, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23517624

RESUMO

Mesenchymal stromal cells (MSCs) participate in repair of damaged tissues, possess the potential to serve as a useful tool in the drug discovery field and exert immunosuppressive effects as demonstrated by their ability to modulate the immune response. Herein, the roles played by MSC differentiation and/or production of trophic factors involved in tissue repair are discussed. MSCs offer the opportunity to probe targets that conventional or differentiated cell lines do not express; thus providing a more refined system that allows identification of novel therapeutics. However, there are difficulties associated with drug discovery assays to which MSCs are not exempt. The immunosuppressive potential of MSCs has already been utilised in clinical trials where MSCs have been used to treat patients with graft- versus- host disease (GvHD) and autoimmune diseases. Another possible therapeutic application of MSCs lies in the field of transplantation tolerance. Although the capacity of MSCs to modulate immune responses has received much attention, the role of MSCs in transplantation tolerance is as yet unclear. In this review, we discuss the evidence for MSC induction of a state of tolerance in the transplantation setting.


Assuntos
Descoberta de Drogas , Tolerância Imunológica , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Cicatrização , Diferenciação Celular , Humanos
8.
Stem Cells Dev ; 22(22): 3003-14, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23795941

RESUMO

Umbilical cord tissue represents a unique source of cells with potential for cell therapy applications for multiple diseases. Human umbilical tissue-derived cells (hUTC) are a developmentally early stage, homogenous population of cells that are HLA-ABC dim, HLA-DR negative, and lack expression of co-stimulatory molecules in the unactivated state. The lack of HLA-DR and co-stimulatory molecule expression on unactivated hUTC may account for their reduced immunogenicity, facilitating their use in allogeneic settings. However, such approaches could be confounded by host innate cells such as natural killer (NK) cells. Here, we evaluate in vitro NK cell interactions with hUTC and compare them with human mesenchymal stem cells (MSC). Our investigations show that hUTC suppress NK activation, through prostaglandin-E2 secretion in a contact-independent manner. Prestimulation of hUTC or human MSC with interferon gamma (IFN-γ) induced expression of the tryptophan degrading enzyme indoleamine 2, 3 dioxygenase, facilitating enhanced suppression. However, resting NK cells of different killer immunoglobulin-like receptor haplotypes did not kill hUTC or MSC; only activated NK cells had the ability to kill nonstimulated hUTC and, to a lesser extent, MSC. The cell killing process involved signaling through the NKG2D receptor and the perforin/granzyme pathway; this was supported by CD54 (ICAM-1) expression by hUTC. IFN-γ-stimulated hUTC or hMSC were less susceptible to NK killing; in this case, protection was associated with elevated HLA-ABC expression. These data delineate the different mechanisms in a two-way interaction between NK cells and two distinct cell therapies, hUTC or hMSC, and how these interactions may influence their clinical applications.


Assuntos
Citotoxicidade Imunológica , Sangue Fetal/efeitos dos fármacos , Interferon gama/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Dinoprostona/imunologia , Dinoprostona/metabolismo , Sangue Fetal/citologia , Sangue Fetal/imunologia , Regulação da Expressão Gênica/imunologia , Granzimas/genética , Granzimas/imunologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Perforina/genética , Perforina/imunologia , Transdução de Sinais
9.
Eur J Immunol ; 41(10): 2840-51, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21710489

RESUMO

Mesenchymal stem cells (MSCs) inhibit T-cell activation and proliferation but their effects on individual T-cell-effector pathways and on memory versus naïve T cells remain unclear. MSC influence on the differentiation of naïve and memory CD4(+) T cells toward the Th17 phenotype was examined. CD4(+) T cells exposed to Th17-skewing conditions exhibited reduced CD25 and IL-17A expression following MSC co-culture. Inhibition of IL-17A production persisted upon re-stimulation in the absence of MSCs. These effects were attenuated when cell-cell contact was prevented. Th17 cultures from highly purified naïve- and memory-phenotype responders were similarly inhibited. Th17 inhibition by MSCs was reversed by indomethacin and a selective COX-2 inhibitor. Media from MSC/Th17 co-cultures contained increased prostaglandin E2 (PGE2) levels and potently suppressed Th17 differentiation in fresh cultures. MSC-mediated Th17 inhibition was reversed by a selective EP4 antagonist and was mimicked by synthetic PGE2 and a selective EP4 agonist. Activation-induced IL-17A secretion by naturally occurring, effector-memory Th17 cells from a urinary obstruction model was also inhibited by MSC co-culture in a COX-dependent manner. Overall, MSCs potently inhibit Th17 differentiation from naïve and memory T-cell precursors and inhibit naturally-occurring Th17 cells derived from a site of inflammation. Suppression entails cell-contact-dependent COX-2 induction resulting in direct Th17 inhibition by PGE2 via EP4.


Assuntos
Dinoprostona/metabolismo , Células-Tronco Mesenquimais/fisiologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/biossíntese , Feminino , Citometria de Fluxo , Indometacina/farmacologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/biossíntese , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Ativação Linfocitária , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Receptores de Prostaglandina E Subtipo EP4/agonistas , Células Th17/efeitos dos fármacos
10.
J Cell Biochem ; 112(8): 1963-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21445861

RESUMO

Adult mesenchymal stem cells possess a remarkably diverse array of immunosuppressive characteristics. The capacity to suppress the regular processes of allogeneic rejection, have allowed the use of tissue mismatched cells as therapeutic approaches in regenerative medicine and as agents of immune deviation. This review describes recent advances in understanding the mechanistic basis of mesenchymal stromal or stem cells (MSC) interaction with innate immunity. Particular emphasis is placed on the effect of Toll-like receptor signalling on MSC and a hypothesis that innate immune signals induce a 'licensing switch' in MSC is put forward. The mechanisms underlying MSC suppression of T cell responses and induction of regulatory populations are surveyed. Conflicting data regarding the influence of MSC on B cell function are outlined and discussed. Finally the limits to MSC mediated immune modulation are discussed with reference to the future clinical application of novel cell therapies.


Assuntos
Células-Tronco Adultas/imunologia , Tolerância Imunológica , Imunidade Inata , Células-Tronco Mesenquimais/imunologia , Células-Tronco Adultas/citologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Humanos , Células-Tronco Mesenquimais/citologia , Transdução de Sinais/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Receptores Toll-Like/imunologia
11.
Hum Gene Ther ; 21(12): 1641-55, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20718666

RESUMO

Allogeneic mesenchymal stem or stromal cells (MSCs) are proposed as cell therapies for degenerative, inflammatory, and autoimmune diseases. The feasibility of allogeneic MSC therapies rests heavily on the concept that these cells avoid or actively suppress the immunological responses that cause rejection of most allogeneic cells and tissues. In this article the validity of the immune privileged status of allogeneic MSCs is explored in the context of recent literature. Current data that provide the mechanistic basis for immune modulation by MSCs are reviewed with particular attention to how MSCs modify the triggering and effector functions of innate and adaptive immunity. The ability of MSCs to induce regulatory dendritic and T-cell populations is discussed with regard to cell therapy for autoimmune disease. Finally, we examine the evidence for and against the immune privileged status of allogeneic MSCs in vivo. Allogeneic MSCs emerge as cells that are responsive to local signals and exert wide-ranging, predominantly suppressive, effects on innate and adaptive immunity. Nonetheless, these cells also retain a degree of immunogenicity in some circumstances that may limit MSC longevity and attenuate their beneficial effects. Ultimately successful allogeneic cell therapies will rely on an improved understanding of the parameters of MSC-immune system interactions in vivo.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Imunidade Inata , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Imunologia de Transplantes
12.
J Gen Virol ; 90(Pt 11): 2622-2633, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19641045

RESUMO

Adeno-associated virus serotype 2 (AAV-2) has been developed as a gene therapy vector. Antibody and cell-mediated immune responses to AAV-2 or AAV-2-transfected cells may confound the therapeutic use of such vectors in clinical practice. In one of the most detailed examinations of AAV-2 immunity in humans to date, cell-mediated and humoral immune responses to AAV-2 were characterized from a panel of healthy blood donors. The extent of AAV-2-specific antibody in humans was determined by examination of circulating AAV-2-specific total IgG levels in plasma from 45 normal donors. Forty-one donors were seropositive and responses were dominated by IgG1 and IgG2 subclasses. Conversely, AAV-2-specific IgG3 levels were consistently low in all donors. Cell-mediated immune recall responses were detectable in nearly half the population studied. In vitro restimulation with AAV-2 of peripheral blood mononuclear cell cultures from 16 donors elicited gamma interferon (IFN-gamma) (ten donors), interleukin-10 (IL-10) (eight donors) and interleukin-13 (IL-13) (four donors) responses. Using a series of overlapping peptides derived from the sequence of the VP1 viral capsid protein, a total of 59 candidate T-cell epitopes were identified. Human leukocyte antigen characterization of donors revealed that the population studied included diverse haplotypes, but that at least 17 epitopes were recognized by multiple donors and could be regarded as immunodominant. These data indicate that robust immunological memory to AAV-2 is established. The diversity of sequences recognized suggests that attempts to modify the AAV-2 capsid, as a strategy to avoid confounding immunity, will not be feasible.


Assuntos
Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Epitopos de Linfócito T/imunologia , Anticorpos Antivirais/sangue , Doadores de Sangue , Células Cultivadas , Variação Genética , Antígenos HLA/genética , Haplótipos , Humanos , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Leucócitos Mononucleares/imunologia
13.
Growth Factors ; 26(1): 12-22, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18365875

RESUMO

Bone morphogenetic proteins (BMPs) are critical morphogens and play key roles in epithelial-mesenchymal transitions (EMTs) during embryogenesis. BMP4 is required for early mesoderm formation and also regulates morphogenesis and epithelial cell differentiation in developing lungs. While, BMP signalling pathways are activated during lung inflammation in adult mice, the role of BMPs in adult lungs remains unclear. We hypothesised that BMPs are involved in remodelling processes in adult lungs and investigated effects of BMP4 on airway epithelial cells. BEAS-2B cell growth decreased in the presence of BMP4. Cells acquired a mesenchymal-like morphology with downregulation of adherens junction proteins and increased cell motility. Changes in extracellular matrix-related gene expression occurred with BMP4 treatment including upregulation of collagens, fibronectin and tenascin C. We conclude that the activity of BMP4 in EMT during development is recapitulated in adult airway epithelial cells and suggest that this activity may contribute to inflammation and fibrosis in vivo.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Células Epiteliais/metabolismo , Epitélio/metabolismo , Regulação da Expressão Gênica , Pulmão/metabolismo , Mesoderma/metabolismo , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/metabolismo , Caderinas/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Citoesqueleto/metabolismo , Humanos , Microscopia de Fluorescência , Modelos Biológicos
14.
Immunol Lett ; 115(1): 50-8, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18022251

RESUMO

Mesenchymal stem cells (MSC) possess a wide range of immunosuppressive functions. Among these is the ability to inhibit CD4+ T cell proliferation. Dendritic cells (DC) play a role in initiating cell-mediated immunity; however, the immunosuppressive influence of MSC on professional antigen presenting cells remains unclear. DC exposed to TNF-alpha and cultured with murine MSC failed to show regular upregulation of maturation markers. Similarly, the presence of MSC abrogated the capacity of ovalbumin-pulsed DC to support antigen specific CD4+ T cell proliferation, or for DC to display an MHC class II- peptide complex recognizable by specific antibody. Interestingly, culture of MSC with DC resulted in reduced expression of CCR7 by DC following stimulation. Likewise, DC matured in the presence of MSC, showed significantly less migration to CCL19. In contrast, murine MSC prevented loss of expression of the tissue anchoring protein E-cadherin by DC. Modulation of DC maturation and function was not permanent and could be restored after removal of MSC. These data demonstrate that MSC modulate the three cardinal features of DC maturation, providing the first demonstration of MSC interference with DC migration.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Quimiotaxia , Células Dendríticas/imunologia , Células-Tronco Mesenquimais/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Quimiocina CCL19/imunologia , Quimiocina CCL19/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Imunidade Celular , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismo
15.
Immunol Lett ; 110(2): 91-100, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17507101

RESUMO

Murine mesenchymal stem cells (MSC) have the ability to inhibit allogeneic immune responses. Two different mechanisms, either cell contact-dependent or independent, have been proposed to account for this immunosuppression. The focus of this study was to elucidate the involvement of soluble suppressive factors secreted by murine MSC in an inflammatory setting, and their role in MSC immunomodulation. In a non-inflammatory environment, bone marrow derived murine MSC constitutively expressed low levels of COX-2, PGE-2, TGF-beta1 and HGF, but not IL-10, PD-1, PD-L1 or PD-L2. These MSC were able to significantly reduce alloantigen driven proliferation in mixed lymphocyte reactions as well as mitogen driven proliferation. The pro-inflammatory cytokines IFN-gamma and TNF-alpha did not ablate MSC mediated immunosuppression. MSC expression of PGE-2, IDO and PD-L1 was differentially regulated by these cytokines. COX-2 and PGE-2 expression by MSC were upregulated by both IFN-gamma and TNF-alpha, and using a biochemical inhibitor this was shown to have an essential, non-redundant role in modulating alloantigen-driven proliferation. However, the surface expression of PD-L1 was induced by IFN-gamma but not TNF-alpha and similarly functional IDO expression was only induced by IFN-gamma stimulation. Blocking studies using neutralising antibodies and biochemical antagonists revealed that while PD-L1 induction was not essential, IDO expression was a prerequisite for IFN-gamma mediated MSC immunomodulation. These data demonstrate that murine MSC expression of immunomodulatory factors dramatically changes in a pro-inflammatory environment and that IFN-gamma in particular has an important role in regulating MSC immunomodulatory factor expression.


Assuntos
Citocinas/imunologia , Interferon gama/imunologia , Células-Tronco Mesenquimais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Células da Medula Óssea , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Feminino , Fatores Imunológicos/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indometacina/farmacologia , Interferon gama/metabolismo , Teste de Cultura Mista de Linfócitos , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Peptídeos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
16.
Immunol Lett ; 103(2): 92-100, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16290206

RESUMO

Mouse models and in vitro cell culture were used to examine airway expression of the mucosal chemokine CCL28. Low levels of constitutively expressed mRNA were observed in transformed murine epithelial cells, but high levels could be induced by stimulation. Cytokines that signal through NF-kappaB, including IL-1beta and TNF-alpha or via JAK-STAT pathway including oncostatin M induced CCL28 in airway epithelial cells in vitro. Immunohistochemistry of murine airway tissue revealed that constitutive expression of CCL28 protein in vivo was low and not ubiquitous. However, abundant expression was detected in epithelia and lymphoid aggregates following allergic sensitization and challenge with ovalbumin. This was accompanied by increased detection of cells expressing CCR10 protein and mRNA in inflamed airways. Taken together, these data support a role for CCL28 in contributing to allergen driven airway pathologies, show that proinflammatory cytokines can induce this signal and suggest a role for CCR10 expressing cells in airway inflammation.


Assuntos
Asma/genética , Quimiocinas CC/genética , Modelos Animais de Doenças , Hipersensibilidade/genética , Receptores CCR10/genética , Animais , Asma/imunologia , Asma/metabolismo , Células Cultivadas , Quimiocinas CC/biossíntese , Células Epiteliais/metabolismo , Feminino , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/metabolismo , Receptores CCR10/biossíntese , Mucosa Respiratória/metabolismo , Regulação para Cima
17.
J Inflamm (Lond) ; 2: 8, 2005 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16045800

RESUMO

Adult bone marrow derived mesenchymal stem cells offer the potential to open a new frontier in medicine. Regenerative medicine aims to replace effete cells in a broad range of conditions associated with damaged cartilage, bone, muscle, tendon and ligament. However the normal process of immune rejection of mismatched allogeneic tissue would appear to prevent the realisation of such ambitions. In fact mesenchymal stem cells avoid allogeneic rejection in humans and in animal models. These finding are supported by in vitro co-culture studies. Three broad mechanisms contribute to this effect. Firstly, mesenchymal stem cells are hypoimmunogenic, often lacking MHC-II and costimulatory molecule expression. Secondly, these stem cells prevent T cell responses indirectly through modulation of dendritic cells and directly by disrupting NK as well as CD8+ and CD4+ T cell function. Thirdly, mesenchymal stem cells induce a suppressive local microenvironment through the production of prostaglandins and interleukin-10 as well as by the expression of indoleamine 2,3,-dioxygenase, which depletes the local milieu of tryptophan. Comparison is made to maternal tolerance of the fetal allograft, and contrasted with the immune evasion mechanisms of tumor cells. Mesenchymal stem cells are a highly regulated self-renewing population of cells with potent mechanisms to avoid allogeneic rejection.

18.
Stem Cells Dev ; 14(3): 252-65, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15969620

RESUMO

Herein we review recent data that support host tolerance of allogeneic adult mesenchymal stem cells (MSC). Evidence is emerging that donor MSC deploy a very powerful array of mechanisms that allow escape from host allogeneic responses. These mechanisms include limited expression of alloantigen by the stem cell and cell contact-dependent and -independent mechanisms. MSC modulate host dendritic cell and T cell function, promoting induction of suppressor or regulatory T cells. These effects are complemented by the induction of divisional arrest anergy in T cells and by stem cell production of soluble immunomodulatory factors, including interleukin-10, transforming growth factor-beta, prostaglandin E2, and hepatocyte growth factor. In addition, MSC express the enzyme indoleamine 2,3-dioxygenase, which creates a tryptophan-depleted milieu that promotes immunosuppression. We propose that these observations show striking similarity to emerging data on the maternal acceptance of the fetal allograft. This comparison suggests new approaches to determine the contribution of different mechanisms to the successful use of MSC in regenerative medicine.


Assuntos
Transplante de Tecido Fetal/imunologia , Mesoderma/imunologia , Células-Tronco/microbiologia , Adulto , Feminino , Rejeição de Enxerto/imunologia , Humanos , Complexo Principal de Histocompatibilidade , Troca Materno-Fetal/imunologia , Gravidez , Transplante Homólogo/imunologia
19.
J Gen Virol ; 86(Pt 7): 1885-1890, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15958666

RESUMO

Infection with influenza virus strongly predisposes an individual to bacterial superinfection, which is often the significant cause of morbidity and mortality during influenza epidemics. Little is known about the immunomodulating properties of the virus that lead to this phenomenon, but the effect of the viral components on the development of immune dendritic cells (DCs) may prove vital. In this study, activation of and cytokine secretion by bacterial lipopolysaccharide (LPS)-stimulated bone marrow-derived dendritic cells (BMDCs) following treatment with the influenza virus major antigen haemagglutinin (HA) were examined. HA selectively inhibits the release of LPS-induced interleukin 12 (IL12) p70, which is independent of IL10 secretion. Suppression occurs at the transcriptional level, with selective inhibition of p35- and not p40-subunit mRNA expression. The downregulation of IL12 p70 by influenza HA is a novel and unexplored pathway that may be relevant in the predisposition to bacterial superinfection associated with influenza virus infections.


Assuntos
Células Dendríticas/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Tolerância Imunológica , Vírus da Influenza A/patogenicidade , Interleucina-12/genética , Interleucina-12/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transcrição Gênica , Animais , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Células Dendríticas/citologia , Regulação para Baixo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Vírus da Influenza A/metabolismo , Subunidade p35 da Interleucina-12 , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
20.
Cell Immunol ; 238(2): 87-96, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16581045

RESUMO

CCL28 is a mucosal chemokine that attracts eosinophils and T cells via the receptors CCR3 and CCR10. Consequently, it is a candidate mediator of the pathology associated with asthma. This study examined constitutive and induced expression of CCL28 by A549 human airway epithelial-like cells. Real-time RT-PCR and ELISA of cultured cells and supernatants revealed constitutive levels of CCL28 expression to be low, whereas IL-1beta and TNF-alpha, induced significantly increased expression. Observations from induced sputum and human airway biopsies supported this. Signal transduction studies revealed that IL-1beta and TNF-alpha stimulation induced NFkappaB phosphorylation in A549 cells, but antagonist inhibition of NFkappaB p50-p65 phosphorylation correlated with marked reduction of IL-1beta or TNF-alpha induced CCL28 expression. Together these studies imply a role for CCL28 in the orchestration of airway inflammation, and suggest that CCL28 is one link between microbial insult and the exacerbation of pathologies such as asthma, through an NFkappaB-dependent mechanism.


Assuntos
Quimiocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Interleucina-1/farmacologia , Pulmão/citologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos , Linhagem Celular Tumoral , Quimiocinas/genética , Quimiocinas CC , Células Epiteliais/metabolismo , Humanos , NF-kappa B/antagonistas & inibidores , Fosforilação , RNA Mensageiro/genética , Escarro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA