Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Trends Mol Med ; 26(3): 296-310, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31862243

RESUMO

New treatments for multiple sclerosis (MS) focused on B cells have created an atmosphere of excitement in the MS community. B cells are now known to play a major role in disease, demonstrated by the highly impactful effect of a B cell-depleting antibody on controlling MS. The idea that a virus may play a role in the development of MS has a long history and is supported mostly by studies demonstrating a link between B cell-tropic Epstein-Barr virus (EBV) and disease onset. Efforts to develop antiviral strategies for treating MS are underway. Although gaps remain in our understanding of the etiology of MS, the role, if any, of viruses in propagating pathogenic immune responses deserves attention.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Animais , Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/virologia , Humanos , Imunoterapia/métodos
2.
J Interferon Cytokine Res ; 35(3): 186-99, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25330068

RESUMO

We demonstrate that interferon (IFN)-ß-1b induces an alternative-start transcript containing the C-terminal TLDc domain of nuclear receptor coactivator protein 7 (NCOA7), a member of the OXR family of oxidation resistance proteins. IFN-ß-1b induces NCOA7-AS (alternative start) expression in peripheral blood mononuclear cells (PBMCs) obtained from healthy individuals and multiple sclerosis patients and human fetal brain cells, astrocytoma, neuroblastoma, and fibrosarcoma cells. NCOA7-AS is a previously undocumented IFN-ß-inducible gene that contains only the last 5 exons of full-length NCOA7 plus a unique first exon (exon 10a) that is not found in longer forms of NCOA7. This exon encodes a domain closely related to an important class of bacterial aldo-keto oxido-reductase proteins that play a critical role in regulating redox activity. We demonstrate that NCOA7-AS is induced by IFN and LPS, but not by oxidative stress and exhibits, independently, oxidation resistance activity. We further demonstrate that induction of NCOA7-AS by IFN is dependent on IFN-receptor activation, the Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling pathway, and a canonical IFN-stimulated response element regulatory sequence upstream of exon 10a. We describe a new role for IFN-ßs involving a mechanism of action that leads to an increase in resistance to inflammation-mediated oxidative stress.


Assuntos
Imunoterapia/métodos , Interferon beta/metabolismo , Leucócitos Mononucleares/fisiologia , Esclerose Múltipla/terapia , Coativadores de Receptor Nuclear/metabolismo , Isoformas de Proteínas/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Humanos , Janus Quinases/metabolismo , Dados de Sequência Molecular , Esclerose Múltipla/imunologia , Coativadores de Receptor Nuclear/genética , Oxirredução , Estresse Oxidativo , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína/genética , Receptores de Interferon/metabolismo , Elementos Reguladores de Transcrição/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
3.
Pancreas ; 44(1): 99-105, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25072284

RESUMO

OBJECTIVES: Interferons (IFNs) have several anticancer mechanisms. A number of clinical trials have been conducted regarding adjuvant IFN-α therapy in pancreatic cancer. Type I IFNs exert their effect via the type I IFN receptor (IFNAR-1, IFNAR-2c). The aims of the present study were to determine the type I IFN receptor expression in pancreatic and periampullary cancer tissues and to study its relation with clinicopathological factors. METHODS: Receptor expression was determined by immunohistochemistry in paraffin-embedded cancer tissue of 47 pancreatic and 54 periampullary cancer patients. RESULTS: The results demonstrated that 91.5% of the pancreatic tumors and 88.9% of the periampullary tumors showed expression of IFNAR-1, of which 23.4% and 13.0% were strongly positive, respectively. Regarding IFNAR-2c expression, 68.1% of the pancreatic tumors and 68.5% of the periampullary tumors were positive, of which 4.3% of the pancreatic tumors and none of the periampullary tumors had a strong expression. No statistically significant associations were found between type I IFN receptor expression and clinicopathological factors or survival. CONCLUSIONS: Type I IFN receptors are expressed in pancreatic and periampullary cancer tissues although with great intertumoral and intratumoral variability. A small proportion of both tumors showed a strong expression of the IFNAR-1; only a very small percentage of the pancreatic tumors showed strong expression of the IFNAR-2c.


Assuntos
Ampola Hepatopancreática/química , Biomarcadores Tumorais/análise , Neoplasias do Ducto Colédoco/química , Neoplasias Pancreáticas/química , Receptor de Interferon alfa e beta/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Ampola Hepatopancreática/patologia , Ampola Hepatopancreática/cirurgia , Neoplasias do Ducto Colédoco/mortalidade , Neoplasias do Ducto Colédoco/patologia , Neoplasias do Ducto Colédoco/terapia , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Fatores de Tempo , Resultado do Tratamento
4.
J Interferon Cytokine Res ; 31(5): 451-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21361792

RESUMO

Synthetic interferons (IFNs) are used in the treatment of several types of cancer. In addition to an antitumor effect, IFNs show antiangiogenic activity. The aim of this study was to investigate the effects of IFN-α and IFN-ß on human micro- and macrovascular endothelial cells in vitro [human micro vascular lung endothelial cells (HMVEC-L) and human umbilical cord endothelial cells (HUVEC)]. By immunohistochemical staining and quantitative reverse transcriptase (RT)-polymerase chain reaction, we studied expression of type I IFN receptors. We evaluated the effects of IFN-α and IFN-ß on the proliferation (DNA content), apoptosis (DNA fragmentation by enzyme-linked immunosorbent assay), and cell cycle distribution (flow-cytometric analysis) of endothelial cells. HUVEC and HMVEC-L cells show comparable expression level of the distinct IFN receptor subtypes. Proliferation of HMVEC-L and HUVEC was inhibited by IFN-ß (the half maximal inhibitory concentration [IC(50)] = 60 and 90 IU/mL, respectively), but not by IFN-α at a dose up to 1,000 IU/mL. An interesting and unexpected observation was an inhibition of apoptosis by IFN-ß. After 72 h of treatment with IFN-ß. Cell cycle inhibition occurs in late S-phase in both cell lines. In conclusion, only IFN-ß, not IFN-α (10-1,000 IU/mL), has an inhibitory activity on endothelial cell proliferation. Surprisingly, apoptosis was decreased by IFN treatment, whereas inhibition of proliferation is caused by cell cycle arrest in late S-phase.


Assuntos
Células Endoteliais/efeitos dos fármacos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon beta/genética , Receptores de Interferon/metabolismo
5.
J Interferon Cytokine Res ; 30(10): 743-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20874251

RESUMO

More than 16 years ago human interferon-β-1b (IFN-β-1β) was shown to be effective in the treatment of the relapsing-remitting form of multiple sclerosis (MS). Over time, IFN-β has been demonstrated to be both a safe and effective treatment. However, the mechanism of action of IFN-β in MS remains unknown. To better understand the mechanism of action of IFN-β, considerable effort has been made in transcriptional profiling of peripheral blood mononuclear cells collected from MS patients. IFN-β is known to induce a large number of genes that play an important role in regulating responses to viral infection, immune modulation, and cell proliferation. Identifying differentially induced genes that are linked to the beneficial effects observed during treatment is under active investigation. IFN biomarkers in MS patients have been proposed but have not been clearly confirmed in independent studies or consistently correlated with clinical measures of disease progression. Organizing single genes or gene signatures grouped according to molecular mechanisms meaningful in MS may help to link IFN activity measurements to clinical outcomes. In this review, IFN activity measurements will be discussed with a specific emphasis on what is known about differential gene expression and treatment effects in MS.


Assuntos
Biomarcadores Tumorais/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Interferon beta/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/genética , Biomarcadores Tumorais/metabolismo , Regulação da Expressão Gênica/genética , Humanos
6.
Inflamm Bowel Dis ; 16(3): 428-41, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19639560

RESUMO

BACKGROUND: Granulocyte-macrophage colony-stimulating factor (GM-CSF) therapy is effective in treating some Crohn's disease (CD) patients and protects mice from colitis induced by dextran sulfate sodium (DSS) administration. However, its mechanisms of action remain elusive. We hypothesized that GM-CSF affects intestinal mucosal repair. METHODS: DSS colitic mice were treated with daily pegylated GM-CSF or saline and clinical, histological, and inflammatory parameters were kinetically evaluated. Further, the role of bone marrow-derived cells in the impact of GM-CSF therapy on DSS colitis was addressed using cell transfers. RESULTS: GM-CSF therapy reduced clinical signs of colitis and the release of inflammatory mediators. GM-CSF therapy improved mucosal repair, with faster ulcer reepithelialization, accelerated hyperproliferative response of epithelial cells in ulcer-adjacent crypts, and lower colonoscopic ulceration scores in GM-CSF-administered mice relative to untreated mice. We observed that GM-CSF-induced promotion of mucosal repair is timely associated with a reduction in neutrophil numbers and increased accumulation of CD11b(+) monocytic cells in colon tissues. Importantly, transfer of splenic GM-CSF-induced CD11b(+) myeloid cells into DSS-exposed mice improved colitis, and lethally irradiated GM-CSF receptor-deficient mice reconstituted with wildtype bone marrow cells were protected from DSS-induced colitis upon GM-CSF therapy. Lastly, GM-CSF-induced CD11b(+) myeloid cells were shown to promote in vitro wound repair. CONCLUSIONS: Our study shows that GM-CSF-dependent stimulation of bone marrow-derived cells during DSS-induced colitis accelerates colonic tissue repair. These data provide a putative mechanism for the observed beneficial effects of GM-CSF therapy in Crohn's disease.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Colite/tratamento farmacológico , Colite/patologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Cicatrização/efeitos dos fármacos , Doença Aguda , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Colite/induzido quimicamente , Colo/efeitos dos fármacos , Colo/patologia , Colo/fisiologia , Doença de Crohn/tratamento farmacológico , Doença de Crohn/patologia , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa/efeitos dos fármacos , Mucosa/patologia , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Organismos Livres de Patógenos Específicos , Cicatrização/fisiologia
7.
J Interferon Cytokine Res ; 30(3): 163-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20038197

RESUMO

Type I interferon (IFN)-dependent STAT1 and STAT2 activation requires specific tyrosine residues (337Y and 512Y) located in the cytoplasmic domain of IFNAR-2c, the beta-subunit of the human type I IFN receptor. To identify STAT activation-independent induction of ISGs, we used a mutant cell line in which both 337Y and 512Y were substituted with phenylalanine (337F512F or FF mutant). In these cells, type I IFN failed to activate STAT1, STAT2, and STAT3 did not induce well-characterized ISGs and did not exert antiviral or antiproliferative effects. Using Oligonucleotide array (Affymetrix) analysis, we showed that interferon regulatory factor-9 (IRF-9) was the only gene induced by IFN-beta in FF cells. Transient transfection analysis using an IRF-9 promoter-reporter luciferase construct in FF cells confirmed induction of the IRF-9 transcription unit by IFN-beta. EMSA analysis using an IFN-stimulated response element (ISRE)-like sequence on the IRF-9 promoter detected 2 novel DNA-binding complexes induced in nuclear extracts of IFN-beta-treated FF cells. Supershift experiments identified the proteins IRF-1 and C/EBP-beta in the complex. These studies provide the first evidence that signaling pathways leading to gene transcription are activated by IFN-beta independent of STAT phosphorylation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon beta/farmacologia , Fatores de Transcrição STAT , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Humanos , Interferon beta/genética , Análise em Microsséries , Dados de Sequência Molecular , Mutação , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais
8.
J Neuroimmunol ; 195(1-2): 116-20, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18279974

RESUMO

The molecular mechanism by which interferon beta (IFN-beta) is effective in treating multiple sclerosis is not well understood. Mononuclear cells from therapy-naïve MS patients, IFN-beta-1b-treated MS patients, and healthy controls were analyzed to examine mRNA changes that characterize both the disease and its treatment. The scientific literature was comprehensively searched for all protein-protein interactions. In MS patients who had never been treated with IFN-beta, statistical analysis revealed coordinate changes in mRNA expression for proteins reported in the literature as "regulated by IFN-beta." As a positive control for this approach, samples from a separate MS patient cohort showed significant change of these same genes during in vivo treatment with IFN-beta-1b.The strength of effect observed for regulation by IFN-beta was greater than for IFN-alpha, IFN-gamma (Th1), or IL-4 (Th2). Of the sets we investigated, the most strongly affected by disease was the subset defined by regulation by both IFN-beta and IFN-alpha. Changes in cells from therapy-naïve MS patients thus anticipated the importance of IFN-beta in therapy. These findings are a significant step towards marrying MS disease etiology and IFN-beta mechanism of action at a molecular level.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Leucócitos Mononucleares/metabolismo , Esclerose Múltipla Recidivante-Remitente/patologia , Estudos de Coortes , Feminino , Humanos , Interferon beta/uso terapêutico , Leucócitos Mononucleares/efeitos dos fármacos , Masculino , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Miastenia Gravis , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fatores de Tempo
9.
Inflamm Bowel Dis ; 14(1): 88-99, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17932977

RESUMO

BACKGROUND: Sargramostim, granulocyte macrophage colony-stimulating factor (GM-CSF), a hematopoietic growth factor, stimulates cells of the intestinal innate immune system. Clinical trials show that sargramostim induces clinical response and remission in patients with active Crohn's disease. To study the mechanism, we examined the effects of GM-CSF in the dextran sulfate sodium (DSS)-induced acute colitis model. We hypothesized that GM-CSF may work through effects on dendritic cells (DCs). METHODS: Acute colitis was induced in Balb/c mice by administration of DSS in drinking water. Mice were treated with daily GM-CSF or phosphate-buffered saline (PBS). To probe the role of plasmacytoid DCs (pDCs) in the response to GM-CSF, we further examined the effects of monoclonal antibody 440c, which is specific for a sialic acid-binding immunoglobulin (Ig)-like lectin expressed on pDCs. RESULTS: GM-CSF ameliorates acute DSS-induced colitis, resulting in significantly improved clinical parameters and histology. Microarray analysis showed reduced expression of proinflammatory genes including TNF-alpha and IL1-beta; the results were further confirmed by real-time reverse-transcriptase polymerase chain reaction and serum Bio-plex analysis. GM-CSF treatment significantly expands pDCs and type 1 IFN production. Administration of mAb 440c completely blocked the therapeutic effect of GM-CSF. GM-CSF is also effective in RAG1(-/-) mice, demonstrating activity-independent effects on T and B cells. IFN-beta administration mimics the therapeutic effect of GM-CSF in DSS-treated mice. GM-CSF increases systemic and mucosal type 1 IFN expression and exhibits synergy with pDC activators, such as microbial cytosine-phosphate-guanosine (CpG) DNA. CONCLUSIONS: GM-CSF is effective in the treatment of DSS colitis in a mechanism involving the 440c(+) pDC population.


Assuntos
Colite/induzido quimicamente , Colite/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Animais , Colite/patologia , Colite/fisiopatologia , Colo/patologia , Células Dendríticas/imunologia , Sulfato de Dextrana/toxicidade , Feminino , Proteínas de Homeodomínio/genética , Interferon Tipo I/imunologia , Interleucina-1beta/genética , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Recombinantes , Fator de Necrose Tumoral alfa/genética
10.
Ann Surg ; 246(2): 259-68, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17667505

RESUMO

OBJECTIVE: We evaluated the role of type I interferons (IFNs) and IFN receptors in the regulation of cell growth in 3 human pancreatic adenocarcinoma cell lines (BxPC-3, MiaPaCa-2, and Panc-1). BACKGROUND: Chemotherapy and radiotherapy have a marginal role in the management of pancreatic adenocarcinoma. The addition of IFN-alpha showed promising results in early clinical trials. METHODS: Cell proliferation and apoptosis were evaluated by DNA measurement and DNA fragmentation, respectively. Type I IFN receptor (IFNAR-1 and IFNAR-2 subunits) was determined by quantitative RT-PCR and immunocytochemistry. Cell cycle distribution was evaluated by propidium iodide staining and flow-cytometric analysis. RESULTS: The incubation with IFN-beta for 6 days showed a potent inhibitory effect on the proliferation of BxPC-3 (IC(50), 14 IU/mL) and MiaPaCa-2 (IC(50), 64 IU/mL). The inhibitory effect of IFN-beta was stronger than IFN-alpha in all 3 cell lines and mainly modulated by the stimulation of apoptosis, although cell cycle arrest was induced as well. The expression of the type I IFN receptors was significantly higher in BxPC-3 (the most sensitive cell line to IFN) and mainly localized on the membrane, whereas in Panc-1 (the most resistant cell line) about 60% to 70% of cells were negative for IFNAR-2c with a mainly cytoplasmic staining for IFNAR-2c. CONCLUSION: The antitumor activity of IFN-beta is more potent than IFN-alpha in pancreatic cancer cell lines through the induction of apoptosis. Further studies should investigate in vivo whether the intensity and distribution of IFNAR-1 and IFNAR-2c may predict the response to therapy with IFN-alpha and IFN-beta in pancreatic cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Interferon Tipo I/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , RNA Mensageiro/genética , Receptor de Interferon alfa e beta/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico , Receptor de Interferon alfa e beta/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
J Clin Endocrinol Metab ; 91(11): 4537-43, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16912135

RESUMO

CONTEXT: Adrenocortical carcinoma (ACC) is a rare tumor with a poor prognosis. Despite efforts to develop new therapeutic regimens for metastatic ACC, surgery remains the mainstay of treatment. Interferons are known to exert tumor-suppressive effects in several types of human cancer. DESIGN: We evaluated the tumor-suppressive effects of type I interferons (IFN)-alpha2b and IFNbeta on the H295 and SW13 human ACC cell lines. RESULTS: As determined by quantitative RT-PCR analysis and immunocytochemistry, H295 and SW13 cells expressed the active type I IFN receptor (IFNAR) mRNA and protein (IFNAR-1 and IFNAR-2c subunits). Both IFNalpha2b and IFNbeta1a significantly inhibited ACC cell growth in a dose-dependent manner, but the effect of IFNbeta1a (IC50 5 IU/ml, maximal inhibition 96% in H295; IC50 18 IU/ml, maximal inhibition 85% in SW13) was significantly more potent, compared with that of IFNalpha2b (IC50 57 IU/ml, maximal inhibition 35% in H295; IC50 221 IU/ml, maximal inhibition 60% in SW13). Whereas in H295 cells both IFNs induced apoptosis and accumulation of the cells in S phase, the antitumor mechanism in SW13 cells involved cell cycle arrest only. Inhibitors of caspase-3, caspase-8, and caspase-9 counteracted the apoptosis-inducing effect by IFNbeta1a in H295 cells. In H295 cells, IFNbeta1a, but not IFNalpha2b, also strongly suppressed the IGF-II mRNA expression, an important growth factor and hallmark in ACC. CONCLUSIONS: IFNbeta1a is much more potent than IFNalpha2b to suppress ACC cell proliferation in vitro by induction of apoptosis and cell cycle arrest. Further studies are required to evaluate the potency of IFNbeta1a to inhibit tumor growth in vivo.


Assuntos
Proliferação de Células/efeitos dos fármacos , Interferon Tipo I/farmacologia , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Carcinoma Adrenocortical/tratamento farmacológico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Humanos , Hidrocortisona/metabolismo , Interferon Tipo I/uso terapêutico , Interferon alfa-2 , Interferon beta-1a , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes , Células Tumorais Cultivadas
12.
Cancer Res ; 66(1): 554-62, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397272

RESUMO

IFN-alpha controls hormone secretion and symptoms in human gastroenteropancreatic neuroendocrine tumors (GEP-NET) but it rarely induces a measurable tumor size reduction. The effect of other type I IFNs, e.g., IFN-beta, has not been evaluated. We compared the antitumor effects of IFN-alpha and IFN-beta in BON cells, a functioning human GEP-NET cell line. As determined by quantitative reverse transcription-PCR analysis and immunocytochemistry, BON cells expressed the active type I IFN receptor mRNA and protein (IFNAR-1 and IFNAR-2c subunits). After 3 and 6 days of treatment, IFN-beta significantly inhibited BON cell growth in a time- and dose-dependent manner. IC50 and maximal inhibitory effect on day 6 were 8 IU/mL and 98%, respectively. In contrast, the effect of IFN-alpha resulted significantly in a less potent effect (IC50: 44 IU/mL, maximal inhibition: 26%). IFN-alpha induced only cell cycle arrest, with an accumulation of the cells in S phase. IFN-beta, apart from a more potent delay in S-G2-M phase transit of the cell cycle, also induced a strong stimulation of apoptosis, evaluated by flow cytometry (Annexin V and 7-AAD) and measurement of the DNA fragmentation. Besides, only IFN-beta severely suppressed chromogranin A levels in the medium from BON cells after 6 days of treatment. In conclusion, IFN-beta is much more potent, compared with IFN-alpha, in its inhibitory effect on GEP-NET cell proliferation in vitro through the induction of apoptosis and cell cycle arrest. Further studies are required to establish whether IFN-beta has comparable potent tumor growth inhibitory effects in vivo.


Assuntos
Tumor Carcinoide/tratamento farmacológico , Neoplasias Gastrointestinais/tratamento farmacológico , Interferon beta/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Tumor Carcinoide/genética , Tumor Carcinoide/metabolismo , Tumor Carcinoide/patologia , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cromogranina A , Cromograninas/biossíntese , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Humanos , Interferon alfa-2 , Interferon beta-1a , Interferon-alfa/farmacologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor de Interferon alfa e beta , Receptores de Interferon/biossíntese , Receptores de Interferon/genética , Proteínas Recombinantes
13.
Int J Cancer ; 111(1): 32-42, 2004 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-15185340

RESUMO

In addition to antiviral effects, Type I interferons (IFN) have potent antiproliferative and immunomodulatory activities. Because of these properties IFNs have been evaluated as therapeutics for the treatment of a number of human diseases, including cancer. Currently, IFNs have been shown to be efficacious for the treatment of only a select number of cancers. The reason for this is unclear. Recent evidence has demonstrated that some cancer cell types seem to be defective in their ability to respond to IFN. It has been suggested that defects in IFN signaling is one mechanism by which cancer cells escape responsiveness to Type I IFNs and growth control in general. We report that transfection and enhanced expression of the Type I IFN receptor chain (IFNAR2c) in 3 different human cancer cell lines markedly increases the sensitivity of these cells to the antiproliferative effects of IFNs. In cancer cells transfected with IFNAR2c, dose response curves demonstrate a significant decrease in the concentrations of IFN required to achieve maximum cell death. Furthermore, in these transfected cells, we observe a significant increase in the number of cells undergoing apoptosis, as measured by DNA fragmentation and Caspase 3 activation. In addition, using an in vivo xenograft tumor model we show an increase in the effectiveness of systemically delivered Betaseron in decreasing tumor burden in animals in which solid tumors were generated from IFNAR2c transfected cells. These data show that specific regulation of IFN receptor expression can play a major role in determining the clinical outcome of IFN-based cancer therapeutics by regulating the relative sensitivity of cancer cells to IFN-dependent growth control.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Interferons/farmacologia , Receptores de Interferon/biossíntese , Animais , Apoptose , Caspase 3 , Caspases/farmacologia , Linhagem Celular Tumoral , Dano ao DNA , Feminino , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Nus , Receptor de Interferon alfa e beta , Receptores de Interferon/fisiologia , Transdução de Sinais , Transfecção , Transplante Heterólogo
14.
J Biol Chem ; 277(38): 35635-41, 2002 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-12105218

RESUMO

Human type I interferons (IFNs) play an important role in the regulation of antiviral defense mechanisms, immunomodulatory activities, and growth control. Recent efforts have demonstrated the importance of IFNs in the activation of signal transducers and activators of transcription (STATs). The role of STAT1 and STAT2 in IFN-dependent JAK-STAT signaling is well established; however, the role of STAT3 and its activation by IFNs remains unclear. Understanding the IFN-dependent regulation of STAT3 is of increasing interest because recent studies have demonstrated that STAT3 may play a role in cancer. Studies have revealed that STAT3 is constitutively active in a number of cancer cell lines and that overexpression of an active form of STAT3 transforms normal fibroblasts. Therefore, STAT3 exhibits properties indicative of known oncogenes. In this report, we define the role of the type I IFN receptor in STAT3 activation and identify for the first time tyrosine residues present in the cytoplasmic domain of IFNAR2c that are critical for STAT3 activation. The regulation of STAT3 activation by IFNs was measured in a human lung fibrosarcoma cell line lacking IFNAR2c but stably expressing various IFNAR2c tyrosine mutants. We show here that in addition to IFN-dependent tyrosine phosphorylation of STAT3, activation using a STAT3-dependent electrophoretic mobility shift assay and a STAT3-specific reporter can also be demonstrated. Furthermore, we demonstrate that type I IFN-dependent activation of STAT3 proceeds through a novel mechanism that is dependent on two tyrosines, Tyr(337) and Tyr(512), present in IFNAR2c and contained within a conserved six-amino acid residue motif, GxGYxM. Surprisingly, both tyrosines were previously shown to be required for type I IFN-dependent STAT1 and STAT2 activation. Our results reveal that type I IFNs activate multiple STATs via the overlapping usage of two tyrosine residues located in the cytoplasmic domain of IFNAR2c.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interferon Tipo I/metabolismo , Receptores de Interferon/metabolismo , Transativadores/metabolismo , Tirosina/metabolismo , Sequência de Bases , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Proteínas de Membrana , Receptor de Interferon alfa e beta , Receptores de Interferon/química , Fator de Transcrição STAT3 , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA