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










Base de dados
Intervalo de ano de publicação
1.
Pathogens ; 11(7)2022 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-35890050

RESUMO

Conventionally, hyperimmune globulin drugs manufactured from pooled immunoglobulins from vaccinated or convalescent donors have been used in treating infections where no treatment is available. This is especially important where multi-epitope neutralization is required to prevent the development of immune-evading viral mutants that can emerge upon treatment with monoclonal antibodies. Using microfluidics, flow sorting, and a targeted integration cell line, a first-in-class recombinant hyperimmune globulin therapeutic against SARS-CoV-2 (GIGA-2050) was generated. Using processes similar to conventional monoclonal antibody manufacturing, GIGA-2050, comprising 12,500 antibodies, was scaled-up for clinical manufacturing and multiple development/tox lots were assessed for consistency. Antibody sequence diversity, cell growth, productivity, and product quality were assessed across different manufacturing sites and production scales. GIGA-2050 was purified and tested for good laboratory procedures (GLP) toxicology, pharmacokinetics, and in vivo efficacy against natural SARS-CoV-2 infection in mice. The GIGA-2050 master cell bank was highly stable, producing material at consistent yield and product quality up to >70 generations. Good manufacturing practices (GMP) and development batches of GIGA-2050 showed consistent product quality, impurity clearance, potency, and protection in an in vivo efficacy model. Nonhuman primate toxicology and pharmacokinetics studies suggest that GIGA-2050 is safe and has a half-life similar to other recombinant human IgG1 antibodies. These results supported a successful investigational new drug application for GIGA-2050. This study demonstrates that a new class of drugs, recombinant hyperimmune globulins, can be manufactured consistently at the clinical scale and presents a new approach to treating infectious diseases that targets multiple epitopes of a virus.

2.
Nat Biotechnol ; 39(8): 989-999, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33859400

RESUMO

Plasma-derived polyclonal antibody therapeutics, such as intravenous immunoglobulin, have multiple drawbacks, including low potency, impurities, insufficient supply and batch-to-batch variation. Here we describe a microfluidics and molecular genomics strategy for capturing diverse mammalian antibody repertoires to create recombinant multivalent hyperimmune globulins. Our method generates of diverse mixtures of thousands of recombinant antibodies, enriched for specificity and activity against therapeutic targets. Each hyperimmune globulin product comprised thousands to tens of thousands of antibodies derived from convalescent or vaccinated human donors or from immunized mice. Using this approach, we generated hyperimmune globulins with potent neutralizing activity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in under 3 months, Fc-engineered hyperimmune globulins specific for Zika virus that lacked antibody-dependent enhancement of disease, and hyperimmune globulins specific for lung pathogens present in patients with primary immune deficiency. To address the limitations of rabbit-derived anti-thymocyte globulin, we generated a recombinant human version and demonstrated its efficacy in mice against graft-versus-host disease.


Assuntos
Linfócitos B/imunologia , COVID-19/terapia , Globulinas/biossíntese , SARS-CoV-2/imunologia , Animais , Anticorpos Antivirais/imunologia , Células CHO , Cricetulus , Ensaio de Imunoadsorção Enzimática , Globulinas/imunologia , Humanos , Imunização Passiva , Camundongos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Zika virus/imunologia , Soroterapia para COVID-19
3.
Stem Cell Res Ther ; 9(1): 95, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29631607

RESUMO

BACKGROUND: Mesenchymal stem cells have potential applications in inflammatory bowel disease due to their immunomodulatory properties. Our aim was to evaluate the feasibility, safety and efficacy of endoscopic administration of adipose-derived mesenchymal stem cells (ASCs) in a colitis model in rats. METHODS: Colitis was induced in rats by rectal trinitrobenzenesulfonic acid (TNBS). After 24 h ASCs (107 cells) or saline vehicle were endoscopically injected into the distal colon. Rats were followed for 11 days. Daily weight, endoscopic score at days 1 and 11, macroscopic appearance at necropsy, colon length and mRNA expression of Foxp3 and IL-10 in mesenteric lymph nodes (MLN) were analyzed. RESULTS: Endoscopic injection was successful in all the animals. No significant adverse events or mortality due to the procedure occurred. Weight evolution was significantly better in the ASC group, recovering initial weight by day 11 (- 0.8% ± 10.1%, mean ± SD), whereas the vehicle group remained in weight loss (- 6.7% ± 9.2%, p = 0.024). The endoscopic score improved in the ASC group by 47.1% ± 5.3% vs. 21.8% ± 6.6% in the vehicle group (p < 0.01). Stenosis was less frequent in the ASC group (4.8% vs. 41.2%, p < 0.01). Colon length significantly recovered in the ASC group versus the vehicle group (222.6 ± 17.3 mm vs. 193.6 ± 17.9 mm, p < 0.001). The endoscopic score significantly correlated with weight change, macroscopic necropsy score and colon length. Foxp3 and IL-10 mRNA levels in MLN recovered with ASC treatment. CONCLUSIONS: ASC submucosal endoscopic injection is feasible, safe and ameliorates TNBS-induced colitis in rats, especially stenosis.


Assuntos
Colite Ulcerativa/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Tecido Adiposo/citologia , Animais , Células Cultivadas , Colite Ulcerativa/etiologia , Colite Ulcerativa/patologia , Constrição Patológica , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Ácido Trinitrobenzenossulfônico/toxicidade
4.
Front Immunol ; 8: 638, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28642759

RESUMO

Mesenchymal stem cells (MSCs) have a large potential in cell therapy for treatment of inflammatory and autoimmune diseases, thanks to their immunomodulatory properties. The encouraging results in animal models have initiated the translation of MSC therapy to clinical trials. In cell therapy protocols with MSCs, administered intravenously, several studies have shown that a small proportion of infused MSCs can traffic to the draining lymph nodes (LNs). This is accompanied with an increase of different types of regulatory immune cells in the LNs, suggesting the importance of migration of MSCs to the LNs in order to contribute to immunomodulatory response. Intranodal (IN), also referred as intralymphatic, injection of cells, like dendritic cells, is being proposed in the clinic for the treatment of cancer and allergy, showing that this route of administration is clinically safe and efficient. In this study, we investigated, for the first time, the biodistribution and the efficacy of Luciferase+ adipose-derived MSCs (Luci-eASCs), infused through the inguinal LNs (iLNs), in normal mice and in inflamed mice with colitis. Most of the Luci-eASCs remain in the iLNs and in the adipose tissue surrounding the inguinal LNs. A small proportion of Luci-eASCs can migrate to other locations within the lymphatic system and to other tissues and organs, having a preferential migration toward the intestine in colitic mice. Our results show that the infused Luci-eASCs protected 58% of the mice against induced colitis. Importantly, a correlation between the response to eASC treatment and a higher accumulation of eASCs in popliteal, parathymic, parathyroid, and mesenteric LNs were found. Altogether, these results suggest that IN administration of eASCs is feasible and may represent an effective strategy for cell therapy protocols with human adipose-derived MSCs in the clinic for the treatment of immune-mediated disorders.

5.
Front Immunol ; 8: 462, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28484460

RESUMO

Mesenchymal stem cells (MSCs) are multipotent stromal cells with immunomodulatory properties. They have emerged as a very promising treatment for autoimmunity and inflammatory diseases such as rheumatoid arthritis. Previous studies have demonstrated that MSCs, administered systemically, migrate to lymphoid tissues associated with the inflammatory site where functional MSC-induced immune cells with a regulatory phenotype were increased mediating the immunomodulatory effects of MSCs. These results suggest that homing of MSCs to the lymphatic system plays an important role in the mechanism of action of MSCs in vivo. Thus, we hypothesized that direct intralymphatic (IL) (also referred as intranodal) administration of MSCs could be an alternative and effective route of administration for MSC-based therapy. Here, we report the feasibility and efficacy of the IL administration of human expanded adipose mesenchymal stem cells (eASCs) in a mouse model of collagen-induced arthritis (CIA). IL administration of eASCs attenuated the severity and progression of arthritis, reduced bone destruction and increased the levels of regulatory T cells (CD25+Foxp3+CD4+ cells) and Tr1 cells (IL10+CD4+), in spleen and draining lymph nodes. Taken together, these results indicate that IL administration of eASCs is very effective in modulating established CIA and may represent an alternative treatment modality for cell therapy with eASCs.

6.
Cytotherapy ; 15(7): 753-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23602595

RESUMO

In the past decade, the therapeutic value of mesenchymal stromal cells (MSCs) has been studied in various indications, thereby taking advantage of their immunosuppressive properties. Easy procurement from bone marrow, adipose tissue or other sources and conventional in vitro expansion culture have made their clinical use attractive. Bridging the gap between current scientific knowledge and regulatory prospects on the transformation potential and possible tumorigenicity of MSCs, the Cell Products Working Party and the Committee for Advanced Therapies organized a meeting with leading European experts in the field of MSCs. This meeting elucidated the risk of potential tumorigenicity related to MSC-based therapies from two angles: the scientific perspective and the regulatory point of view. The conclusions of this meeting, including the current regulatory thinking on quality, nonclinical and clinical aspects for MSCs, are presented in this review, leading to a clearer way forward for the development of such products.


Assuntos
Carcinogênese , Proliferação de Células , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Diferenciação Celular/genética , Humanos , Células-Tronco Mesenquimais/metabolismo
7.
Tissue Eng Part A ; 18(7-8): 852-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22059379

RESUMO

Human adipose-derived stem cells (hASC) are mesenchymal stem cells with reduced immunogenicity and the ability to modulate immune responses. APRIL and BAFF proteins are overexpressed in inflammatory and autoimmune diseases for which allogeneic hASC therapy is currently under clinical investigation. Modification of hASC properties by the tissue microenvironment could be a critical factor in patient outcome and is still not well understood. Our aim was to characterize the APRIL/BAFF system in hASC by analyzing the ligand and receptor expression patterns, the effects mediated by APRIL and BAFF on hASC, and the underlying signaling. We found that hASC express the tumor necrosis factor proteins APRIL (a proliferation-inducing ligand) and BAFF (B cell-activator factor) as well as their receptors TACI (transmembrane activator and calcium-modulator and cyclophilin ligand interactor), BCMA (B cell maturation antigen) and the BAFF-specific receptor (BAFF-R). APRIL and BAFF secretion was differentially enhanced by CXCL12 and interferon (IFN)-γ, implicated in hASC-mediated migration and immunosuppression, respectively. In addition, APRIL and BAFF induced rapid phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2) and Akt kinases and promoted an increase in hASC proliferation, without affecting the immunosuppressive capacity of these cells. The use of specific chemical inhibitors indicated that the PI3K transduction pathway is involved in hASC basal growth and that APRIL- and BAFF-mediated effects are ERK-dependent. These results provide new information about the molecular mechanisms that underlie APRIL and BAFF secretion and signaling in hASC, and are of special relevance for the use of allogeneic hASC as therapeutic tools.


Assuntos
Tecido Adiposo/citologia , Fator Ativador de Células B/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Adulto , Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/metabolismo , Antígeno de Maturação de Linfócitos B/genética , Antígeno de Maturação de Linfócitos B/metabolismo , Proliferação de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Interleucina-6/metabolismo , Interleucina-8 , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
8.
J Mol Cell Cardiol ; 49(5): 771-80, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20713059

RESUMO

Myocardial infarction caused by vascular occlusion results in the formation of nonfunctional fibrous tissue. Cumulative evidence indicates that cell therapy modestly improves cardiac function; thus, novel cell sources with the potential to repair injured tissue are actively sought. Here, we identify and characterize a cell population of cardiac adipose tissue-derived progenitor cells (ATDPCs) from biopsies of human adult cardiac adipose tissue. Cardiac ATDPCs express a mesenchymal stem cell-like marker profile (strongly positive for CD105, CD44, CD166, CD29 and CD90) and have immunosuppressive capacity. Moreover, cardiac ATDPCs have an inherent cardiac-like phenotype and were able to express de novo myocardial and endothelial markers in vitro but not to differentiate into adipocytes. In addition, when cardiac ATDPCs were transplanted into injured myocardium in mouse and rat models of myocardial infarction, the engrafted cells expressed cardiac (troponin I, sarcomeric α-actinin) and endothelial (CD31) markers, vascularization increased, and infarct size was reduced in mice and rats. Moreover, significant differences between control and cell-treated groups were found in fractional shortening and ejection fraction, and the anterior wall remained significantly thicker 30days after cardiac delivery of ATDPCs. Finally, cardiac ATDPCs secreted proangiogenic factors under in vitro hypoxic conditions, suggesting a paracrine effect to promote local vascularization. Our results indicate that the population of progenitor cells isolated from human cardiac adipose tissue (cardiac ATDPCs) may be valid candidates for future use in cell therapy to regenerate injured myocardium.


Assuntos
Tecido Adiposo/citologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Idoso , Indutores da Angiogênese/metabolismo , Animais , Capilares/patologia , Diferenciação Celular , Linhagem da Célula , Separação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Testes de Função Cardíaca , Humanos , Camundongos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Neovascularização Fisiológica , Ratos , Ultrassonografia
9.
J Neurosci ; 30(4): 1502-11, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107078

RESUMO

Transforming growth factors-beta (TGF-betas) signal through type I and type II serine-threonine kinase receptor complexes. During ligand binding, type II receptors recruit and phosphorylate type I receptors, triggering downstream signaling. BAMBI [bone morphogenetic protein (BMP) and activin membrane-bound inhibitor] is a transmembrane pseudoreceptor structurally similar to type I receptors but lacks the intracellular kinase domain. BAMBI modulates negatively pan-TGF-beta family signaling; therefore, it can be used as an instrument for unraveling the roles of these cytokines in the adult CNS. BAMBI is expressed in regions of the CNS involved in pain transmission and modulation. The lack of BAMBI in mutant mice resulted in increased levels of TGF-beta signaling activity, which was associated with attenuation of acute pain behaviors, regardless of the modality of the stimuli (thermal, mechanical, chemical/inflammatory). The nociceptive hyposensitivity exhibited by BAMBI(-/-) mice was reversed by the opioid antagonist naloxone. Moreover, in a model of chronic neuropathic pain, the allodynic responses of BAMBI(-/-) mice also appeared attenuated through a mechanism involving delta-opioid receptor signaling. Basal mRNA and protein levels of precursor proteins of the endogenous opioid peptides proopiomelanocortin (POMC) and proenkephalin (PENK) appeared increased in the spinal cords of BAMBI(-/-). Transcript levels of TGF-betas and their intracellular effectors correlated directly with genes encoding opioid peptides, whereas BAMBI correlated inversely. Furthermore, incubation of spinal cord explants with activin A or BMP-7 increased POMC and/or PENK mRNA levels. Our findings identify TGF-beta family members as modulators of acute and chronic pain perception through the transcriptional regulation of genes encoding the endogenous opioids.


Assuntos
Vias Aferentes/metabolismo , Proteínas de Membrana/metabolismo , Nervos Periféricos/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Medula Espinal/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Proteína Morfogenética Óssea 7/metabolismo , Proteína Morfogenética Óssea 7/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Encefalinas/genética , Encefalinas/metabolismo , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Antagonistas de Entorpecentes/farmacologia , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Nervos Periféricos/fisiopatologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/fisiopatologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/fisiopatologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Regulação para Cima/genética
10.
Arthritis Rheum ; 60(4): 1006-19, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19333946

RESUMO

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic autoimmune disease caused by loss of immunologic self tolerance and characterized by chronic joint inflammation. Adult mesenchymal stem cells (MSCs) were recently found to suppress effector T cell responses and to have beneficial effects in various immune disorders. The purpose of this study was to examine a new therapeutic strategy for RA based on the administration of human adipose-derived MSCs (AD-MSCs). METHODS: DBA/1 mice with collagen-induced arthritis were treated with human AD-MSCs after disease onset, and clinical scores were determined. Inflammatory response was determined by measuring the levels of different mediators of inflammation in the joints and serum. The Th1-mediated autoreactive response was evaluated by determining the proliferative response and cytokine profile of draining lymph node cells stimulated with the autoantigen. The number of Treg cells and the suppressive capacity on self-reactive Th1 cells were also determined. RESULTS: Systemic infusion of human AD-MSCs significantly reduced the incidence and severity of experimental arthritis. This therapeutic effect was mediated by down-regulating the 2 deleterious disease components: the Th1-driven autoimmune and inflammatory responses. Human AD-MSCs decreased the production of various inflammatory cytokines and chemokines, decreased antigen-specific Th1/Th17 cell expansion, and induced the production of antiinflammatory interleukin-10 in lymph nodes and joints. Human AD-MSCs also induced de novo generation of antigen-specific CD4+CD25+FoxP3+ Treg cells with the capacity to suppress self-reactive T effector responses. CONCLUSION: Human AD-MSCs emerge as key regulators of immune tolerance by inducing the generation/activation of Treg cells and are thus attractive candidates for a cell-based therapy for RA.


Assuntos
Tecido Adiposo/citologia , Artrite Experimental/terapia , Artrite Reumatoide/terapia , Tolerância Imunológica/imunologia , Transplante de Células-Tronco Mesenquimais , Animais , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Antígenos CD4/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Transplante Heterólogo
11.
Tissue Eng Part A ; 15(10): 2795-806, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19231921

RESUMO

Human adipose-derived mesenchymal stem cells (hASCs) are mesenchymal stem cells (MSCs) with reduced immunogenicity and capability to modulate immune responses. Whereas the immunosuppressive activity of bone marrow-MSCs has received considerable attention during the last few years, the specific mechanisms underlying hASC-mediated immunosuppression have been poorly studied. Recent studies comparing both cell types have reported differences at transcriptional and proteomic levels, suggesting that hASCs and bone marrow-MSCs, while having similarities, are quite different. This suggests that different mechanisms of immunosuppression may apply. Here, we report that hASCs inhibit peripheral blood mononuclear cells (PBMCs), and CD4(+) and CD8(+) T cell proliferation in both cell-cell contact and transwell conditions, which is accompanied by a reduction of proinflammatory cytokines. We demonstrate that hASCs do not constitutively express immunomodulatory factors. Conditioned supernatants from hASCs stimulated by IFN-gamma, PBMCs, or activated PBMCs highly inhibited PBMC proliferation, indicating that inhibitory factors are released upon hASC activation. Many factors have been involved in MSC-mediated immunosuppression, including IFN-gamma, IL-10, hepatocyte growth factor, prostaglandin E2, transforming growth factor-beta1, indoleamine 2,3-dioxygenase (IDO), nitric oxide, and IL-10. Using pharmacological inhibitors, neutralizing antibodies, and genetically modified hASCs that constitutively express or silence IDO enzyme, we demonstrate that, in the case of hASCs, the IFN-gamma/IDO axis is essential. Taken together, our data support the key role of IDO in the therapeutic use of hASC on immunomediated diseases.


Assuntos
Adipócitos/citologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Linfócitos/citologia , Células-Tronco Mesenquimais/citologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Dinoprostona/metabolismo , Citometria de Fluxo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Interleucina-10/metabolismo , Leucócitos Mononucleares/citologia , Linfócitos/imunologia , Óxido Nítrico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Engenharia Tecidual , Fator de Crescimento Transformador beta1/metabolismo
12.
Gastroenterology ; 136(3): 978-89, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19135996

RESUMO

BACKGROUND & AIMS: Crohn's disease is a chronic disease characterized by severe T-helper (Th)1 cell-driven inflammation of the colon partially caused by a loss of immune tolerance against mucosal antigens. Mesenchymal stem cells were recently described to suppress effector T-cell responses and have therapeutic effects in some immune disorders. Here, we investigated the potential therapeutic effects of human adipose-derived mesenchymal stem cells (hASCs) in a model of inflammatory bowel disease. METHODS: Mice with trinitrobenzene sulfonic acid-induced colitis were treated with hASCs after onset of disease and clinical scores were evaluated. Inflammatory response was determined by measuring the levels of different inflammatory mediators in colon and serum. Th1-mediated effector responses were evaluated by determining the proliferation and cytokine profile of activated mesenteric lymph node cells. The number of regulatory T cells and the suppressive capacity on Th1 cell responses was determined. RESULTS: Systemic infusion of hASCs or murine ASCs ameliorated the clinical and histopathologic severity of colitis, abrogating body weight loss, diarrhea, and inflammation and increasing survival (P < .001). This therapeutic effect was mediated by down-regulating both Th1-driven autoimmune and inflammatory responses. ASCs decreased a wide panel of inflammatory cytokines and chemokines and increased interleukin-10 levels (P < .001), directly acting on activated macrophages. hASCs also impaired Th1 cell expansion and induced/activated CD4(+)CD25(+)FoxP3(+) regulatory T cells with suppressive capacity on Th1 effector responses in vitro and in vivo (P < .001). CONCLUSIONS: hASCs emerge as key regulators of immune tolerance and as attractive candidates for a cell-based therapy for Crohn's disease.


Assuntos
Tecido Adiposo/citologia , Doença de Crohn/imunologia , Doença de Crohn/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Autoimunidade , Células Cultivadas , Colite/imunologia , Colite/patologia , Colite/terapia , Doença de Crohn/patologia , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia , Células Th1/imunologia
13.
Tissue Eng Part A ; 15(7): 1579-89, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19061425

RESUMO

Human adipose-derived stem cells (hASCs) are mesenchymal stem cells with reduced immunogenicity and the capability to modulate immune responses. These properties make hASCs of special interest as therapeutic agents in the settings of chronic inflammatory and autoimmune diseases. Exogenous and endogenous toll-like receptor (TLR) ligands have been linked with the perpetuation of inflammation in a number of chronic inflammatory diseases such as inflammatory bowel disease and rheumatoid arthritis because of the permanent exposure of the immune system to TLR-specific stimuli. Therefore, hASCs employed in therapy are potentially exposed to TLR ligands, which may result in the modulation of hASC activity and therapeutic potency. In this study, we demonstrate that hASCs possess active TLR2, TLR3, and TLR4, because activation with specific ligands resulted in induction of nuclear factor kappa B-dependent genes, such as manganese superoxide dismutase and the release of interleukin (IL)-6 and IL-8. TLR3 and TLR4 ligands increased osteogenic differentiation, but no effect on adipogenic differentiation or proliferation was observed. Moreover, we show that TLR activation does not impair the immunogenic and immunosuppressive properties of hASCs. These results may have important implications with respect to the safety and efficacy of hASC-based cell therapies.


Assuntos
Adipócitos/citologia , Tolerância Imunológica/imunologia , Transdução de Sinais , Células-Tronco/imunologia , Receptores Toll-Like/metabolismo , Adulto , Diferenciação Celular , Proliferação de Células , Feminino , Regulação da Expressão Gênica , Humanos , Fatores Imunológicos/metabolismo , Ligantes , Masculino , Fenótipo , Células-Tronco/citologia , Receptores Toll-Like/genética
14.
Development ; 135(9): 1589-95, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18356246

RESUMO

Although the T-box family of transcription factors function in many different tissues, their role in liver development is unknown. Here we show that Tbx3, the T-box gene that is mutated in human ulnar-mammary syndrome, is specifically expressed in multipotent hepatic progenitor cells, ;hepatoblasts', isolated from the developing mouse liver. Tbx3-deficient hepatoblasts presented severe defects in proliferation as well as uncontrollable hepatobiliary lineage segregation, including the promotion of cholangiocyte (biliary epithelial cell) differentiation, which thereby caused abnormal liver development. Deletion of Tbx3 resulted in the increased expression of the tumor suppressor p19(ARF) (Cdkn2a), which in turn induced a growth arrest in hepatoblasts and activated a program of cholangiocyte differentiation. Thus, Tbx3 plays a crucial role in controlling hepatoblast proliferation and cell-fate determination by suppressing p19(ARF) expression and thereby promoting liver organogenesis.


Assuntos
Diferenciação Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Fígado/citologia , Células-Tronco Multipotentes/citologia , Proteínas com Domínio T/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Fígado/embriologia , Camundongos , Células-Tronco Multipotentes/fisiologia
15.
PLoS One ; 2(4): e398, 2007 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-17460765

RESUMO

BACKGROUND: The heart forms from a linear tube that is subject to complex remodeling during embryonic development. Hallmarks of this remodeling are the looping of the heart tube and the regionalization into chamber and non-chamber myocardium. Cardiomyocytes in the future chamber myocardium acquire different cellular and physiological characteristics through activation of a chamber-specific genetic program, which is in part mediated by T-box genes. METHODOLOGY/PRINCIPAL FINDING: We characterize two new zebrafish T-box transcription factors, tbx3b and tbx2a, and analyze their role during the development of the atrioventricular canal. Loss- and gain-of-function analyses demonstrate that tbx3b and tbx2a are necessary to repress the chamber-genetic program in the non-chamber myocardium. We also show that tbx3b and tbx2a are required to control cell proliferation in the atrioventricular canal and that misregulation of cell proliferation in the heart tube influences looping. Furthermore, we characterize the heart phenotype of a novel Tbx3 mutation in mice and show that both the control of cell proliferation and the repression of chamber-specific genetic program in the non-chamber myocardium are conserved roles of Tbx3 in this species. CONCLUSIONS/SIGNIFICANCE: Taken together, our results uncover an evolutionarily conserved role of Tbx2/3 transcription factors during remodeling of the heart myocardium and highlight the importance of controlling cell proliferation as a driving force of morphogenesis.


Assuntos
Proliferação de Células , Coração/fisiologia , Fatores de Transcrição/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/genética , Peixe-Zebra
16.
Nature ; 427(6970): 121-8, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14712268

RESUMO

During vertebrate embryo development, the breaking of the initial bilateral symmetry is translated into asymmetric gene expression around the node and/or in the lateral plate mesoderm. The earliest conserved feature of this asymmetric gene expression cascade is the left-sided expression of Nodal, which depends on the activity of the Notch signalling pathway. Here we present a mathematical model describing the dynamics of the Notch signalling pathway during chick embryo gastrulation, which reveals a complex and highly robust genetic network that locally activates Notch on the left side of Hensen's node. We identify the source of the asymmetric activation of Notch as a transient accumulation of extracellular calcium, which in turn depends on left-right differences in H+/K+-ATPase activity. Our results uncover a mechanism by which the Notch signalling pathway translates asymmetry in epigenetic factors into asymmetric gene expression around the node.


Assuntos
Padronização Corporal , Sinalização do Cálcio , Ácido Egtázico/análogos & derivados , Proteínas de Membrana/metabolismo , Fatores de Transcrição , Animais , Proteínas Aviárias , Padronização Corporal/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Embrião de Galinha , Ácido Egtázico/farmacologia , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Proteínas de Membrana/genética , Modelos Biológicos , Proteína Nodal , Omeprazol/farmacologia , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Notch1 , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Notch , Proteínas Serrate-Jagged , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
17.
Proc Natl Acad Sci U S A ; 100 Suppl 1: 11889-95, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12909711

RESUMO

Several vertebrates display the ability to regenerate parts of their body after amputation. During this process, differentiated cells reenter the cell cycle and proliferate to generate a mass of undifferentiated cells. Repatterning mechanisms act on these cells to eventually shape a regenerated tissue or organ that replaces the amputated one. Experiments with regenerating limbs/fins in newts and zebrafish have shown that members of the Msx family of homeodomain-containing transcription factors play key roles during blastema formation and patterning. Here we show that adult zebrafish have a remarkable capacity to regenerate the heart in a process that involves up-regulation of msxB and msxC genes. We present evidence indicating that heart regeneration involves the execution of a specific genetic program, rather than redeployment of a cardiac development program. Preceding Msx activation, there is a marked increase in the expression of notch1b and deltaC, which we show are also up-regulated during fin regeneration. These data suggest a role for the Notch pathway in the activation of the regenerative response. Taken together, our results underscore the use of zebrafish as a model for investigating the process of regeneration in particular and the biology of stem cells in general. Advances in these fields will undoubtedly aid in the implementation of strategies for regenerative medicine.


Assuntos
Coração/fisiologia , Proteínas de Membrana/fisiologia , Regeneração , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Coração/crescimento & desenvolvimento , Proteínas de Homeodomínio/genética , Hibridização In Situ , Proteínas de Membrana/genética , Modelos Biológicos , Receptores Notch , Regeneração/genética , Regeneração/fisiologia , Transdução de Sinais , Fatores de Transcrição/genética , Regulação para Cima , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Nat Cell Biol ; 5(6): 513-9, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12766772

RESUMO

The mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and phosphatidylinositol-3-OH kinase (PI3K)/Akt pathways are involved in the regulatory mechanisms of several cellular processes including proliferation, differentiation and apoptosis. Here we show that during chick, mouse and zebrafish limb/fin development, a known MAPK/ERK regulator, Mkp3, is induced in the mesenchyme by fibroblast growth factor 8 (FGF8) signalling, through the PI3K/Akt pathway. This correlates with a high level of phosphorylated ERK in the apical ectodermal ridge (AER), where Mkp3 expression is excluded. Conversely, phosphorylated Akt is detected only in the mesenchyme. Constitutively active Mek1, as well as the downregulation of Mkp3 by small interfering RNA (siRNA), induced apoptosis in the mesenchyme. This suggests that MKP3 has a key role in mediating the proliferative, anti-apoptotic signalling of AER-derived FGF8.


Assuntos
Extremidades/embriologia , Fatores de Crescimento de Fibroblastos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais , Animais , Apoptose , Embrião de Galinha , Fosfatase 6 de Especificidade Dupla , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Ativação Enzimática , Fator 8 de Crescimento de Fibroblasto , Fatores de Crescimento de Fibroblastos/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Dados de Sequência Molecular , Morfogênese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Peixe-Zebra
19.
Genes Dev ; 17(10): 1213-8, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12730123

RESUMO

Left-sided expression of Nodal in the lateral plate mesoderm is a conserved feature necessary for the establishment of normal left-right asymmetry during vertebrate embryogenesis. By using gain- and loss-of-function experiments in zebrafish and mouse, we show that the activity of the Notch pathway is necessary and sufficient for Nodal expression around the node, and for proper left-right determination. We identify Notch-responsive elements in the Nodal promoter, and unveil a direct relationship between Notch activity and Nodal expression around the node. Our findings provide evidence for a mechanism involving Notch activity that translates an initial symmetry-breaking event into asymmetric gene expression.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/fisiologia , Fator de Crescimento Transformador beta/genética , Animais , Elementos Facilitadores Genéticos , Proteínas Hedgehog , Camundongos , Proteína Nodal , Organizadores Embrionários/fisiologia , Regiões Promotoras Genéticas , Receptores Notch , Transdução de Sinais/fisiologia , Situs Inversus/embriologia , Transativadores/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
20.
Development ; 129(22): 5161-70, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12399308

RESUMO

A major gap in our knowledge of development is how the growth and identity of tissues and organs are linked during embryogenesis. The vertebrate limb is one of the best models to study these processes. Combining mutant analyses with gain- and loss-of-function approaches in zebrafish and chick embryos, we show that Tbx5, in addition to its role governing forelimb identity, is both necessary and sufficient for limb outgrowth. We find that Tbx5 functions downstream of WNT signaling to regulate Fgf10, which, in turn, maintains Tbx5 expression during limb outgrowth. Furthermore, our results indicate that Tbx5 and Wnt2b function together to initiate and specify forelimb outgrowth and identity. The molecular interactions governed by members of the T-box, Wnt and Fgf gene families uncovered in this study provide a framework for understanding not only limb development, but how outgrowth and identity of other tissues and organs of the embryo may be regulated.


Assuntos
Extremidades/embriologia , Fatores de Crescimento de Fibroblastos/genética , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas com Domínio T/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Embrião de Galinha , Embrião não Mamífero , Fator 10 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/metabolismo , Botões de Extremidades/fisiologia , Dados de Sequência Molecular , Mutação , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Proteínas Wnt , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...