RESUMO
Intercellular communication through the secretion of soluble factors plays a vital role in a wide range of biological processes (e.g., homeostasis, immune response), yet identification and quantification of many of these factors can be challenging due to their degradation or sequestration in cell culture media prior to analysis. Here, we present a customizable bead-based system capable of simultaneously binding to live cells (through antibody-mediated cell tethering) and capturing cell-secreted molecules. Our functionalized beads capture secreted molecules (e.g., hepatocyte growth factor secreted by fibroblasts) that are diminished when sampled via traditional supernatant analysis techniques (p < 0.05), effectively rescuing a reduced signal in the presence of neutralizing components in the cell culture media. Our system enables capture and analysis of molecules integral to chemical communication that would otherwise be markedly decreased prior to analysis.
Assuntos
Antígenos de Superfície/metabolismo , Fator de Crescimento de Hepatócito/análise , Microesferas , Anticorpos/imunologia , Antígenos de Superfície/análise , Antígenos de Superfície/imunologia , Linhagem Celular , Meios de Cultura/química , Fibroblastos/citologia , Fibroblastos/metabolismo , Fluoresceínas/química , Fator de Crescimento de Hepatócito/imunologia , Fator de Crescimento de Hepatócito/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoensaio/métodos , Antígenos Thy-1/análise , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismoRESUMO
: YYB-101 is a humanized rabbit anti-human hepatocyte growth factor (HGF)-neutralizing antibody currently in clinical trial. To test the effect of HGF neutralization with antibody on anti-cancer T cell immunity, we generated surrogate antibodies that are reactive to the mouse homologue of the epitope targeted by YYB-101. First, we immunized a chicken with human HGF and monitored changes in the B cell repertoire by next-generation sequencing (NGS). We then extracted the VH gene repertoire from the NGS data, clustered it into components by sequence homology, and classified the components by the change in the number of unique VH sequences and the frequencies of the VH sequences within each component following immunization. Those changes should accompany the preferential proliferation and somatic hypermutation or gene conversion of B cells encoding HGF-reactive antibodies. One component showed significant increases in the number and frequencies of unique VH sequences and harbored genes encoding antibodies that were reactive to human HGF and competitive with YYB-101 for HGF binding. Some of the antibodies also reacted to mouse HGF. The selected VH sequences shared 98.3% identity and 98.9% amino acid similarity. It is therefore likely that the antibodies encoded by them all react to the epitope targeted by YYB-101.
Assuntos
Anticorpos/genética , Anticorpos/imunologia , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Fator de Crescimento de Hepatócito/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Sequência de Aminoácidos , Animais , Anticorpos/química , Linfócitos B/imunologia , Linfócitos B/metabolismo , Galinhas , Mapeamento de Epitopos/métodos , Biblioteca Gênica , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , CamundongosRESUMO
BACKGROUND: Immunosuppression following lung transplantation is a key aspect to the graft's survival. However, the well-known complications that are caused by immunosuppressive regimens present an opportunity to study ways to minimize the usage of these drugs. Recently, a promising discovery has been made pertaining to the immunomodulatory effects of adipose tissue-derived mesenchymal stem cells (ADMSCs) through their secretion of hepatocyte growth factor. In the hopes of mitigating the adverse effects of standard immunosuppressive regimens, our study aims to investigate the effects of ADMSCs on the immune response utilizing a rat lung transplantation model. METHODS: Each rat's own ADMSCs were intravenously administered immediately after orthotopic left lung transplantation. The experimental subjects were divided into four groups: 1) control group (group C) was administered no treatment following transplantation; 2) ADMSC group (group A), administered a single intravenous injection of ADMSCs following transplantation; 3) tacrolimus group (group T), administered tacrolimus (0.5 mg/kg) every 24 h following transplantation; and 4) ADMSC and tacrolimus group (AT group) administered a single intravenous injection of ADMSCs in combination with tacrolimus every 24 h following transplantation. RESULTS: The histologically proven rejection grade in group AT was significantly lower than that in group T. The serum levels of hepatocyte growth factor and the expression of cMet in group AT accompanied by low CD40 expression were also significantly higher than those of the lung grafts of group T. CONCLUSIONS: These results suggest that co-administration of ADMSCs with tacrolimus is a beneficial therapeutic approach in lung transplantation.
Assuntos
Tecido Adiposo/citologia , Rejeição de Enxerto/prevenção & controle , Imunoterapia/métodos , Transplante de Pulmão/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Administração Intravenosa , Animais , Terapia Combinada/métodos , Modelos Animais de Doenças , Rejeição de Enxerto/sangue , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Fator de Crescimento de Hepatócito/sangue , Fator de Crescimento de Hepatócito/imunologia , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Pulmão/patologia , Pulmão/cirurgia , Masculino , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Endogâmicos Lew , Tacrolimo/administração & dosagem , Tacrolimo/efeitos adversos , Transplante Autólogo/métodos , Resultado do TratamentoRESUMO
MST1 deficiency causes T and B cell lymphopenia, resulting in combined immunodeficiency. However, MST1-deficient patients also exhibit autoimmune-like symptoms such as hypergammaglobulinemia and autoantibody production. Recent studies have shown that the autoimmune responses observed in MST1-deficient patients were most likely attributable to defective regulatory T (Treg) cells instead of intrinsic signals in MST1-lacking B cells. Nevertheless, it is not determined how MST1 deficiency in T cells breaks B cell tolerance and causes systemic autoimmune-like phenotypes. In this study, we confirmed that Mst1-/- mice developed hypergammaglobulinemia associated with increased levels of IgG, IgA, and IgE. We also showed that uncontrolled B cell responses were resulted from the IL-4-rich environment created by CD4+ T cells. Defective MST1-FOXO1 signaling down-regulated Treg cells, resulting in the collapse of immune tolerance where the populations of Th2 and T follicular helper cells expanded. In conclusion, we suggest that MST1 acts as a molecular brake to maintain immune tolerance by regulating T cell-mediated B cell activation.
Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Fator de Crescimento de Hepatócito/imunologia , Hipergamaglobulinemia/imunologia , Interleucina-4/imunologia , Proteínas Proto-Oncogênicas/imunologia , Animais , Fator de Crescimento de Hepatócito/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/deficiênciaRESUMO
Hepatocyte growth factor (HGF) is a pleiotropic cytokine composed of an α-chain and a ß-chain, and these chains contain four kringle domains and a serine protease-like structure, respectively. The receptor for HGF was identified as the c-met proto-oncogene product of transmembrane receptor tyrosine kinase. HGF-induced signaling through the receptor Met provokes dynamic biological responses that support morphogenesis, regeneration, and the survival of various cells and tissues, which includes hepatocytes, renal tubular cells, and neurons. Characterization of tissue-specific Met knockout mice has further indicated that the HGF-Met system modulates immune cell functions and also plays an inhibitory role in the progression of chronic inflammation and fibrosis. However, the biological actions that are driven by the HGF-Met pathway all play a role in the acquisition of the malignant characteristics in tumor cells, such as invasion, metastasis, and drug resistance in the tumor microenvironment. Even though oncogenic Met signaling remains the major research focus, the HGF-Met axis has also been implicated in infectious diseases. Many pathogens try to utilize host HGF-Met system to establish comfortable environment for infection. Their strategies are not only simply change the expression level of HGF or Met, but also actively hijack HGF-Met system and deregulating Met signaling using their pathogenic factors. Consequently, the monitoring of HGF and Met expression, along with real-time detection of Met activation, can be a beneficial biomarker of these infectious diseases. Preclinical studies designed to address the therapeutic significance of HGF have been performed on injury/disease models, including acute tissue injury, chronic fibrosis, and cardiovascular and neurodegenerative diseases. Likewise, manipulating the HGF-Met system with complete control will lead to a tailor made treatment for those infectious diseases.
Assuntos
Fator de Crescimento de Hepatócito/imunologia , Fator de Crescimento de Hepatócito/fisiologia , Infecções/imunologia , Infecções/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Bactérias/imunologia , Bactérias/metabolismo , Bactérias/patogenicidade , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/uso terapêutico , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met/deficiência , Proteínas Proto-Oncogênicas c-met/genética , Transdução de Sinais , Vírus/imunologia , Vírus/metabolismo , Vírus/patogenicidadeRESUMO
c-Met is a prototypic member of a sub-family of RTKs. Inappropriate c-Met activation plays a crucial role in tumor formation, proliferation and metastasis. Using a key c-Met dimerization assay, a set of 12 murine whole IgG1 monoclonal antibodies was selected and a lead candidate, m224G11, was humanized by CDR-grafting and engineered to generate a divalent full antagonist humanized IgG1 antibody, hz224G11. Neither m224G11 nor hz224G11 bind to the murine c-Met receptor. Their antitumor activity was investigated in vitro in a set of experiments consistent with the reported pleiotropic effects mediated by c-Met and, in vivo, using several human tumor xenograft models. Both m224G11 and hz224G11 exhibited nanomolar affinities for the receptor and inhibited HGF binding, c-Met phosphorylation, and receptor dimerization in a similar fashion, resulting in a profound inhibition of all c-Met functions in vitro. These effects were presumably responsible for the inhibition of c-Met's major functions including cell proliferation, migration, invasion scattering, morphogenesis and angiogenesis. In addition to these in vitro properties, hz224G11 dramatically inhibits the growth of autocrine, partially autophosphorylated and c-Met amplified cell lines in vivo. Pharmacological studies performed on Hs746T gastric cancer xenografts demonstrate that hz224G11 strongly downregulates c-Met expression and phosphorylation. It also decreases the tumor mitotic index (Ki67) and induces apoptosis. Taken together, the in vitro and in vivo data suggest that hz224G11 is a promising candidate for the treatment of tumors. This antibody, now known as ABT-700 and currently in Phase I clinical trials, may provide a novel therapeutic approach to c-Met-expressing cancers.
Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Murinos/farmacologia , Neoplasias/terapia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/imunologia , Células A549 , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Murinos/imunologia , Células CHO , Linhagem Celular Tumoral , Cricetulus , Feminino , Fator de Crescimento de Hepatócito/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoglobulina G/imunologia , Ligantes , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias/imunologia , Proteínas Proto-Oncogênicas c-met/biossíntese , Distribuição Aleatória , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Hepatocyte growth factor (HGF) signaling via the MET receptor is essential for embryonic development and tissue repair. On the other hand, deregulated MET signaling promotes tumor progression in diverse types of cancers. Even though oncogenic MET signaling remains the major research focus, the HGF-MET axis has also been implicated in diverse aspects of immune cell development and functions. In the presence of other hematopoietic growth factors, HGF promotes the development of erythroid, myeloid and lymphoid lineage cells and thrombocytes. In monocytes and macrophages responding to inflammatory stimuli, induction of autocrine HGF-MET signaling can contribute to tissue repair via stimulating anti-inflammatory cytokine production. HGF-MET signaling can also modulate adaptive immune response by facilitating the migration of Langerhans cells and dendritic cells to draining lymph nodes. However, MET signaling has also been shown to induce tolerogenic dendritic cells in mouse models of graft-versus-host disease and experimental autoimmune encephalomyelitis. HGF-MET axis is also implicated in promoting thymopoiesis and the survival and migration of B lymphocytes. Recent studies have shown that MET signaling induces cardiotropism in activated T lymphocytes. Further understanding of the HGF-MET axis in the immune system would allow its therapeutic manipulation to improve immune cell reconstitution, restore immune homeostasis and to treat immuno-inflammatory diseases.
Assuntos
Linfócitos B/imunologia , Fator de Crescimento de Hepatócito/imunologia , Proteínas Proto-Oncogênicas c-met/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Movimento Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Humanos , CamundongosRESUMO
Suppressor of cytokine signaling 1 (SOCS1) is an indispensable regulator of IFNγ signaling and has been implicated in the regulation of liver fibrosis. However, it is not known whether SOCS1 mediates its anti-fibrotic functions in the liver directly, or via modulating IFNγ, which has been implicated in attenuating hepatic fibrosis. Additionally, it is possible that SOCS1 controls liver fibrosis by regulating hepatic stellate cells (HSC), a key player in fibrogenic response. While the activation pathways of HSCs have been well characterized, the regulatory mechanisms are not yet clear. The goals of this study were to dissociate IFNγ-dependent and SOCS1-mediated regulation of hepatic fibrogenic response, and to elucidate the regulatory functions of SOCS1 in HSC activation. Liver fibrosis was induced in Socs1(-/-)Ifng(-/-) mice with dimethylnitrosamine or carbon tetrachloride. Ifng(-/-) and C57BL/6 mice served as controls. Following fibrogenic treatments, Socs1(-/-)Ifng(-/-) mice showed elevated serum ALT levels and increased liver fibrosis compared to Ifng(-/-) mice. The latter group showed higher ALT levels and fibrosis than C57BL/6 controls. The livers of SOCS1-deficient mice showed bridging fibrosis, which was associated with increased accumulation of myofibroblasts and abundant collagen deposition. SOCS1-deficient livers showed increased expression of genes coding for smooth muscle actin, collagen, and enzymes involved in remodeling the extracellular matrix, namely matrix metalloproteinases and tissue inhibitor of metalloproteinases. Primary HSCs from SOCS1-deficient mice showed increased proliferation in response to growth factors such as HGF, EGF and PDGF, and the fibrotic livers of SOCS1-deficient mice showed increased expression of the Pdgfb gene. Taken together, these data indicate that SOCS1 controls liver fibrosis independently of IFNγ and that part of this regulation may occur via regulating HSC proliferation and limiting growth factor availability.
Assuntos
Fator de Crescimento Epidérmico/imunologia , Fator de Crescimento de Hepatócito/imunologia , Interferon gama/imunologia , Cirrose Hepática/imunologia , Fator de Crescimento Derivado de Plaquetas/imunologia , Transdução de Sinais/imunologia , Proteína 1 Supressora da Sinalização de Citocina/imunologia , Animais , Fator de Crescimento Epidérmico/genética , Fator de Crescimento de Hepatócito/genética , Interferon gama/genética , Cirrose Hepática/genética , Cirrose Hepática/patologia , Camundongos , Camundongos Knockout , Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/genética , Proteína 1 Supressora da Sinalização de Citocina/genéticaRESUMO
Human Wharton's Jelly-derived Mesenchymal Stem Cells (hWJ-MSCs) are considered as an alternative for bone-marrow-derived MSCs. These cells have immunosuppressive properties. It was unclear whether the WJ-MSCs would sustain their immunomodulatory characteristics after lentiviral transduction or not. In this study, we evaluated immunomodulatory properties of WJ-MSCs after lentiviral transduction. HWJ-MSCs were transduced with lentiviral particles. Expression of transduced and un-transduced hWJ-MSCs surface molecules and secretion of IL-10, HGF, VEGF and TGF-ß was analyzed. Cell proliferation and frequency of CD4(+)CD25(+) CD127(low/neg) Foxp3(+) T regulatory cells was measured. There was no difference between the surface markers and secretion of IL-10, HGF, VEGF and TGF-ß in transduced and un-transduced hWJ-MSCs. Both cells inhibited the proliferation of PHA stimulated PBMCs, and improved the frequency of T regulatory cells. These findings suggest that lentiviral transduction does not alter the immunomodulatory function of hWJ-MSCs. However, lentiviral transduction may have a wide range of applications in gene therapy.
Assuntos
Diferenciação Celular/imunologia , Fatores Imunológicos/imunologia , Células-Tronco Mesenquimais/imunologia , Geleia de Wharton/citologia , Feminino , Citometria de Fluxo , Fator de Crescimento de Hepatócito/análise , Fator de Crescimento de Hepatócito/imunologia , Humanos , Fatores Imunológicos/genética , Interleucina-10/análise , Interleucina-10/imunologia , Lentivirus/genética , Leucócitos Mononucleares , Células-Tronco Mesenquimais/citologia , Gravidez , Transdução Genética , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/imunologia , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/imunologia , Geleia de Wharton/imunologiaRESUMO
Pulmonary silicosis is an irreversible and untreatable disease that is characterized by interstitial lesions and perpetual fibrosis in the lungs. This study was performed to determine whether mesenchymal stem cells (MSCs) and hepatocyte growth factor (HGF) could exhibit therapeutic effects on human silicosis. This non-randomized uncontrolled trial comprised four patients with pulmonary silicosis who had developed lung fibrosis and received autologous bone marrow MSCs previously transfected by a vector containing human HGF cDNA (MSCs/HGF). MSCs/HGF were intravenously administered weekly for three consecutive weeks at a dose of 2 x 10(6) cells/kg. Pulmonary function, high kilo-voltage chest X-ray radiography, computed tomography (CT) scan, and peripheral blood lymphocyte subset and serum IgG concentrations were evaluated after cell therapy. The treatment was found to be generally safe. Symptoms such as cough and chest distress gradually ameliorated at six months post-therapy, accompanied by the significant improvement of pulmonary function. The ratios of the peripheral CD4- and CD8- positive cell concentrations were increased (P < 0.05). Furthermore, the serum IgG levels in these patients were decreased and reached the normal range (P < 0.05). CT scans showed partial absorption of the nodular and reticulonodular lesions in the lungs during follow-up of at least 12 months. The effectiveness of this novel regimen observed in these patients suggests that a placebo-controlled clinical trial needs to be developed. This study carries trial registration No. NCT01977131 (ClinicalTrials.gov).
Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Fator de Crescimento de Hepatócito/genética , Transplante de Células-Tronco Mesenquimais , Fibrose Pulmonar/terapia , Silicose/terapia , Administração Intravenosa , Adulto , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Relação CD4-CD8 , Feminino , Seguimentos , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Fator de Crescimento de Hepatócito/imunologia , Humanos , Imunoglobulina G/sangue , Pulmão/imunologia , Pulmão/patologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Fibrose Pulmonar/sangue , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Testes de Função Respiratória , Silicose/sangue , Silicose/imunologia , Silicose/patologia , Transfecção , Transplante Autólogo , Resultado do TratamentoRESUMO
We mapped the cytokine response to hematopoietic stem cell transplantation (HSCT) by assaying 51 cytokines and chemokines each week for 100 days in 51 children receiving allogeneic (n = 44) or autologous HSCT (n = 7). Assay values were reported as mean fluorescence intensity (MFI). Log transformation converted MFI to clinically relevant measures (ie, pg/mL). We searched for potential markers of transplant complications by using mixed treatment by subject analysis of variance. Global cytokine secretion in HSCT recipients was significantly lower than in concurrent control patients (n = 11). Coincident with the nadir in WBC count, the concentration of many cytokines declined further by the second and third week. All analytes (except monokine induced by gamma interferon [MIG]) subsequently rebounded by week 4 (coincident with engraftment and recovery of WBC count) but often still remained well below control levels. Concurrent with the collective nadir of multiple cytokines, monocyte chemoattractant protein 1 (MCP-1), growth-regulated oncogene alpha (GRO-a), and leptin surged during weeks 2 to 4. High levels of leptin persisted throughout the 100 post-transplant days. Also during weeks 2 to 4, hepatocyte growth factor (HGF) and IL-6 surged in children with complications but not in those without complications. The peak in HGF was more pronounced in veno-occlusive disease (VOD). HGF and IL-6 secretion rose at least 2 weeks before the clinical diagnosis of VOD or graft-versus-host disease (GVHD). From week 4 onward in all groups, the MFI of the cytokine resistin increased to 5 to 15 times above concurrent control. HGF has now emerged in 3 or more biomarker discovery efforts for GVHD (and in our population for VOD as well). HGF (with or without IL-6) should be investigated as a potential predictive biomarker of VOD or GVHD. Alternatively, the hyperinflammatory "signature" provided by a multicytokine assay may be predictive.
Assuntos
Antineoplásicos/uso terapêutico , Citocinas/imunologia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Fator de Crescimento de Hepatócito/imunologia , Resistina/imunologia , Adolescente , Criança , Pré-Escolar , Citocinas/metabolismo , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Hepatopatia Veno-Oclusiva/imunologia , Hepatopatia Veno-Oclusiva/patologia , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Lactente , Recém-Nascido , Fígado/irrigação sanguínea , Fígado/imunologia , Fígado/patologia , Masculino , Estudos Prospectivos , Resistina/metabolismo , Análise de Sobrevida , Transplante Autólogo , Transplante HomólogoRESUMO
TAK-701 is a humanized antibody that binds hepatocyte growth factor (HGF), thus suppressing c-Met transduced signaling and c-Met dependent proliferation and migration of tumor cells. Six childhood solid tumor xenografts were selected for evaluating TAK-701 based on immunochemical detection of HGF/c-Met autocrine signaling [i.e., pMet(Tyr1349) and HGF positive]. TAK-701 was tested using a dose of 30 mg/kg administered by the intraperitoneal (IP) route twice weekly for 4 weeks. TAK-701 did not induce significant differences in EFS distribution in treated tumors compared to control tumors. Objective responses were not observed in any of the tested solid tumor xenografts.
Assuntos
Anticorpos Monoclonais/uso terapêutico , Glioblastoma/tratamento farmacológico , Fator de Crescimento de Hepatócito/imunologia , Neuroblastoma/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Rabdomiossarcoma Embrionário/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Proliferação de Células/efeitos dos fármacos , Criança , Feminino , Glioblastoma/imunologia , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuroblastoma/imunologia , Neuroblastoma/metabolismo , Osteossarcoma/imunologia , Osteossarcoma/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-met/imunologia , Rabdomiossarcoma Embrionário/imunologia , Rabdomiossarcoma Embrionário/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
AIMS: To define if there is an imbalance in plasma levels of proinflammatory, fibrogenic and antifibrogenic cytokines in patients with liver cirrhosis (LC) and impaired glucose tolerance (IGT) or diabetes mellitus (DM). MATERIAL AND METHODS: We randomly selected 54 out of 100 patients with LC who had normal fasting plasma glucose (FPG) levels. Three groups were formed based on an oral glucose tolerance test (OGTT) results: 18 patients were normal, 18 had IGT, and 18 had DM. Plasma levels of cytokines were measured: TNF- α, soluble tumor necrosis factor receptor 1 (sTNF-R1), leptin, TGF-ß1, and hepatocyte growth factor (HGF). Also, fasting plasma insulin (FPI) levels were determined and HOMA2-IR was calculated. Results were compared with those of a control group of 18 patients without liver disease nor DM. Intergroup comparison was performed using non parametric tests. RESULTS: Significantly higher sTNF-R1 and lower TGF-ß1 were found in patients with IGT and DM compared to controls. Leptin, HGF, and TNF-α levels showed no significant differences. According to Child-Pugh classification all cytokines levels were impaired in groups B or C as compared to group A. Positive correlations between sTNF-R1 and HOMA2-IR and between leptin and HOMA2-IR were found. CONCLUSIONS: IGT and DM were associated with abnormalities of sTNF-R1 and TGF-ß1 compared to non cirrhotic controls. Among cirrhotic patients impairment of all cytokines were more marked in advanced liver disease. Finally, sTNF-R1 and leptin correlated with IR. These findings suggest that IGT and DM may be causally implicated with liver inflammation process.
Assuntos
Citocinas/imunologia , Diabetes Mellitus Tipo 2/imunologia , Intolerância à Glucose/imunologia , Insulina/sangue , Cirrose Hepática/imunologia , Adulto , Idoso , Glicemia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/complicações , Teste de Tolerância a Glucose , Fator de Crescimento de Hepatócito/imunologia , Humanos , Resistência à Insulina , Leptina/imunologia , Cirrose Hepática/sangue , Cirrose Hepática/complicações , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Fator de Crescimento Transformador beta1/imunologia , Fator de Necrose Tumoral alfa/imunologia , Adulto JovemRESUMO
BACKGROUND: Accumulating evidence suggests a deleterious role for CD8+ T cells in multiple sclerosis (MS) pathogenesis. We have recently reported that hepatocyte growth factor (HGF), a potent neuroprotective factor, limits CD4+ T cell-mediated autoimmune neuroinflammation by promoting tolerogenic dendritic cells (DCs) and subsequently regulatory T cells. Whether HGF modulates cell-mediated immunity driven by MHC class I-restricted CD8+ T cells remains to be determined. METHODS: Here we examined whether HGF regulates antigen-specific CD8+ T cell responses using an established model of murine cytotoxic T lymphocyte (CTL)-mediated killing. RESULTS: We found that HGF treatment of gp100-pulsed DCs reduced the activation of gp100-specific T cell receptor (Pmel-1) CD8+ T cells and subsequent MHC class I-restricted CTL-mediated cytolysis of gp100-pulsed target cells. The levels of perforin, granzyme B, IFN-γ, and the degranulation marker CD107a as well as Fas ligand were decreased among CD8+ T cells, suggestive of a dual inhibitory effect of HGF on the perforin/granzyme B- and Fas-based lytic pathways in cell-mediated cytotoxicity. Treatment of CD8+ T cells with concanamycin A, a potent inhibitor of the perforin-mediated cytotoxic pathway, abrogated CTL cytotoxicity indicating that blockade of the perforin-dependent killing is a major mechanism by which HGF diminished cytolysis of gp100-pulsed target cells. Moreover, HGF suppressed the generation of effector memory CTLs. CONCLUSIONS: Our findings indicate that HGF treatment limits both the generation and activity of effector CTL from naïve CD8+ T cells. Complementary to its impact on CD4+ T-cell CNS autoimmunity and myelin repair, our findings further suggest that HGF treatment could be exploited to control CD8+ T-cell-mediated, MHC I-restricted autoimmune dysfunctions such as MS.
Assuntos
Citotoxicidade Imunológica/imunologia , Fator de Crescimento de Hepatócito/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , CamundongosRESUMO
The characterization of the humoral response in cancer patients is becoming a practical alternative to improve early detection. We prepared phage microarrays containing colorectal cancer cDNA libraries to identify phage-expressed peptides recognized by tumor-specific autoantibodies from patient sera. From a total of 1536 printed phages, 128 gave statistically significant values to discriminate cancer patients from control samples. From this, 43 peptide sequences were unique following DNA sequencing. Six phages containing homologous sequences to STK4/MST1, SULF1, NHSL1, SREBF2, GRN, and GTF2I were selected to build up a predictor panel. A previous study with high-density protein microarrays had identified STK4/MST1 as a candidate biomarker. An independent collection of 153 serum samples (50 colorectal cancer sera and 103 reference samples, including healthy donors and sera from other related pathologies) was used as a validation set to study prediction capability. A combination of four phages and two recombinant proteins, corresponding to MST1 and SULF1, achieved an area under the curve of 0.86 to correctly discriminate cancer from healthy sera. Inclusion of sera from other different neoplasias did not change significantly this value. For early stages (A+B), the corrected area under the curve was 0.786. Moreover, we have demonstrated that MST1 and SULF1 proteins, homologous to phage-peptide sequences, can replace the original phages in the predictor panel, improving their diagnostic accuracy.
Assuntos
Autoanticorpos/imunologia , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/imunologia , Fator de Crescimento de Hepatócito/imunologia , Análise Serial de Proteínas/métodos , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Sulfotransferases/imunologia , Bacteriófago T7/imunologia , Neoplasias Colorretais/sangue , Neoplasias Colorretais/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Mutagênese Insercional , Proteínas de Neoplasias/metabolismo , Peptídeos/metabolismo , Curva ROC , Reprodutibilidade dos TestesRESUMO
Hepcidin is a key regulator of iron metabolism. The expression of hepcidin is significantly induced by iron overload, inflammation, and infection of pathogens. Recent studies have indicated that the expression of hepcidin in the liver is also regulated during liver regeneration. However, the mechanism of the regulation of hepcidin expression and its role in liver regeneration remain unclear. In this study, we found that the hepatocyte growth factor inhibited hepcidin expression in the liver during the late stage of liver regeneration. Meanwhile, we investigated the effect of hepcidin on liver regeneration. Mice overexpressing hepcidin-1 exhibited impaired hepatic regeneration after partial hepatectomy, as determined by immunohistochemical staining of the proliferation cell nuclear antigen. Our results demonstrated a negative role of hepcidin in modulating liver regeneration, and suggested that a sustained high iron level by the down-regulation of hepcidin at the late stage of liver regeneration is required for hepatocyte proliferation.
Assuntos
Expressão Gênica/genética , Hepatectomia/métodos , Hepcidinas/genética , Regeneração Hepática/genética , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Proliferação de Células , Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/sangue , Fator de Crescimento de Hepatócito/imunologia , Hepatócitos/citologia , Hepatócitos/metabolismo , Hepcidinas/fisiologia , Imuno-Histoquímica , Interleucina-6/sangue , Ferro/sangue , Regeneração Hepática/fisiologia , Masculino , Camundongos Endogâmicos BALB C , Antígeno Nuclear de Célula em Proliferação/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de TempoRESUMO
Immune-mediated diseases of the CNS, such as multiple sclerosis and its animal model, experimental autoimmune encephalitis (EAE), are characterized by the activation of antigen-presenting cells and the infiltration of autoreactive lymphocytes within the CNS, leading to demyelination, axonal damage, and neurological deficits. Hepatocyte growth factor (HGF) is a pleiotropic factor known for both neuronal and oligodendrocytic protective properties. Here, we assess the effect of a selective overexpression of HGF by neurons in the CNS of C57BL/6 mice carrying an HGF transgene (HGF-Tg mice). EAE induced either by immunization with myelin oligodendrocyte glycoprotein peptide or by adoptive transfer of T cells was inhibited in HGF-Tg mice. Notably, the level of inflammatory cells infiltrating the CNS decreased, except for CD25(+)Foxp3(+) regulatory T (T(reg)) cells, which increased. A strong T-helper cell type 2 cytokine bias was observed: IFN-gamma and IL-12p70 decreased in the spinal cord of HGF-Tg mice, whereas IL-4 and IL-10 increased. Antigen-specific response assays showed that HGF is a potent immunomodulatory factor that inhibits dendritic cell (DC) function along with differentiation of IL-10-producing T(reg) cells, a decrease in IL-17-producing T cells, and down-regulation of surface markers of T-cell activation. These effects were reversed fully when DC were pretreated with anti-cMet (HGF receptor) antibodies. Our results suggest that, by combining both potentially neuroprotective and immunomodulatory effects, HGF is a promising candidate for the development of new treatments for immune-mediated demyelinating diseases associated with neurodegeneration such as multiple sclerosis.
Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Fator de Crescimento de Hepatócito/imunologia , Tolerância Imunológica , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno , Proliferação de Células , Citocinas/biossíntese , Citocinas/imunologia , Encefalomielite Autoimune Experimental/genética , Fatores de Transcrição Forkhead/imunologia , Fator de Crescimento de Hepatócito/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Baço/citologia , Baço/imunologia , Linfócitos T Reguladores/citologia , Células Th2/imunologiaRESUMO
The limited success of dendritic cell (DC)-based immunotherapy in multiple myeloma is partly due to hepatocyte growth factor (HGF)-induced DC dysfunction. From a therapeutic standpoint, it is important to understand the molecular events involved in inhibition of DC activation/maturation by HGF. Because Bruton's tyrosine kinase (Btk) negatively regulates maturation and immunostimulatory function of DCs, a role for Btk in HGF-induced inhibition of both murine and human DCs was investigated. We demonstrate that Btk is a novel proximal component of HGF-induced c-MET (HGF receptor) signaling. Following HGF treatment, Btk binds to c-MET and becomes activated. Btk activation in turn blocks the NF-κB pathway and subsequent DC activation via the c-Src-PI3K-AKT-mammalian target of rapamycin (mTOR) pathway. Notably, Btk activation is necessary for HGF-induced association of c-Src and PI3K with c-MET. Furthermore, we provide the first evidence that HGF inhibits DC activation by inducing autocrine interleukin (IL)-10 secretion, which requires activation of Btk. Blocking activation of Btk and its downstream the c-Src-PI3K-AKT-mTOR pathway prevents HGF-induced IL-10 secretion by DCs. In addition, neutralization of IL-10 secretion from DCs impaired the inhibitory effect of HGF on DCs. Thus, our study identifies a novel role for Btk in HGF-induced DC inhibition.
Assuntos
Células Dendríticas/imunologia , Fator de Crescimento de Hepatócito/imunologia , Proteínas Tirosina Quinases/imunologia , Tirosina Quinase da Agamaglobulinemia , Animais , Proteína Tirosina Quinase CSK , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Ativação Enzimática/imunologia , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Interleucina-10/imunologia , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/imunologia , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-met/imunologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo , Quinases da Família srcRESUMO
Mesenchymal stem cells derived from human umbilical cord Wharton's jelly (hWJMSCs) became prospective seed cell candidate for tissue engineering and cell-based therapy because of its variety source, easy procurement, robust proliferation, and high purity compared with bone marrow- and adipose-derived MSCs. Such neonatal stem cells can be isolated from a variety of extraembryonic tissues and appear to be more primitive and have greater multi-potentiality than their adult counterparts. In this study, we investigated the immune characters of hWJMSCs and its derived cartilage cells (hWJMSC-Cs) by detecting the expression of major histocompatibility complex I/I(MHC-I/II), costimulatory molecules (CD40, CD80 and CD86) and immune inhibitors including human leukocyte antigen G (HLA-G), indoleamine-2,3-dioxygenase (IDO), and prostaglandin E2 (PGE2). We found that hWJMSCs did not express MHC-II and costimulatory molecules, but moderately expressed MHC-I, and positively expressed immune inhibitors as HLA-G, IDO, PGE2, demonstrating their very low immunogenicity and potential to induce immune tolerance microenvironment in hosts. The results of chondrogenic differentiated hWJMSCs(hWJMSC-Cs) are similar to those of undifferentiated cells, except for the slightly elevated MHC-II and costimulators expression. Additionally, we detected cytokine profile of hWJMSCs through cytokine antibody array and verified by western blot the positive expression of immune suppression-related molecules, HGF, VEGF, TGF, and IL-10. Furthermore, to investigate the in vivo immune response of the cells, hWJMSCs-scaffold constructs were implanted into rabbits and rats, and the result showed that hWJMSCs did not elicit immune rejection in the animals. Their intermediate state between adult and embryonic stem cells makes them an ideal candidate for reprogramming to the pluripotent status. Additional studies are necessary to clarify the potential of hWJMSCs to be used in cartilage and other tissue regeneration and cell-based therapies.
Assuntos
Cartilagem/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Cordão Umbilical/imunologia , Geleia de Wharton/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Cartilagem/citologia , Cartilagem/metabolismo , Diferenciação Celular , Dinoprostona/genética , Dinoprostona/imunologia , Expressão Gênica , Antígenos HLA-G/genética , Antígenos HLA-G/imunologia , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/imunologia , Humanos , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Coelhos , Ratos , Transdução de Sinais , Fatores de Crescimento Transformadores/genética , Fatores de Crescimento Transformadores/imunologia , Transplante Heterólogo , Cordão Umbilical/citologia , Cordão Umbilical/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Geleia de Wharton/citologia , Geleia de Wharton/metabolismoRESUMO
BACKGROUND: The c-Met receptor tyrosine kinase is aberrantly activated in many solid tumors. In a prior study we showed that prostate cancer PC-3 cells exhibit constitutively activated c-Met without exogenous hepatocyte growth factor (HGF); however whether this characteristic is due to an endogenous HGF/c-Met autocrine loop remains controversial. In the current study we examined the response of PC-3 cells to an anti-HGF neutralizing antibody or a small molecule Met kinase inhibitor (BMS-777607). METHODS: Cell scattering was tested by monitoring cell morphology after HGF stimulation. Cell migration was examined by both "wound-healing" and transwell assasy and invasion was detected by Matrigel-coated transwell assay. Proliferation, survival and anoikis were determined by MTT, colony formation and trypan blue exclusion assay, respectively. Gene and protein expression were assessed by real-time PCR and Western blot, respectively. RESULTS: Although HGF mRNA could be detected in PC-3 cells, the molecular weight of secreted "HGF" protein was inconsistent with the functional recombinant HGF. Furthermore, conditioned medium from PC-3 cell cultures was ineffective at triggering either motogenic behavior or c-Met signaling in DU145, another prostate cancer cell line expressing c-Met but lacking basal c-Met activation. PC-3 cells also were not responsive to the anti-HGF neutralizing antibody in experiments assessing proliferation, migration, or c-Met signaling. BMS-777607 treatment with micromolar doses nonetheless led to significant inhibition of multiple PC-3 cell functions including proliferation, clonogenicity, migration and invasion. At the molecular level, BMS-777607 suppressed autophosphorylated c-Met and downstream c-Src and Akt pathways. CONCLUSIONS: These results suggest that the constitutive c-Met activation in PC-3 is independent of autocrine stimulation. Because PC-3 cells were responsive to BMS-777607 but not the anti-HGF antibody, the findings also indicate that under circumstances where c-Met is constitutively hyperactive in the absence of functional HGF, targeting the c-Met receptor remains a viable therapeutic option to impede cancer progression.