Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 9(8)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34413169

RESUMO

BACKGROUND: We performed a clinical trial to evaluate safety and immunogenicity of a novel long peptide vaccine administered in combinations of incomplete Freund's adjuvant (IFA) and agonists for TLR3 (polyICLC) and TLR7/8 (resiquimod). We hypothesized that T cell responses to minimal epitope peptides (MEPs) within the long peptides would be enhanced compared with prior vaccines with MEP themselves and that T cell responses would be enhanced with TLR agonists, compared with IFA alone. METHODS: Participants with resected stage IIB-IV melanoma were vaccinated with seven long melanoma peptides (LPV7) from tyrosinase, gp100, MAGE-A1, MAGE-A10, and NY-ESO-1, each containing a known MEP for CD8+ T cells, plus a tetanus helper peptide (Tet) restricted by Class II MHC. Enrollment was guided by an adaptive design to one of seven adjuvant combinations. Vaccines were administered at weeks 1, 2, 3, 6, 9, 12 at rotating injection sites. T cell and IgG antibody (Ab) responses were measured with IFN-gamma ELIspot assay ex vivo and ELISA, respectively. RESULTS: Fifty eligible participants were assigned to seven study groups, with highest enrollment on arm E (LPV7+Tet+IFA+polyICLC). There was one dose-limiting toxicity (DLT) in Group E (grade 3 injection site reaction, 6% DLT rate). All other treatment-related adverse events were grades 1-2. The CD8+ T cell immune response rate (IRR) to MEPs was 18%, less than in prior studies using MEP vaccines in IFA. The CD8+ T cell IRR trended higher for IFA-containing adjuvants (24%) than adjuvants containing only TLR agonists (6%). Overall T cell IRR to full-length LPV7 was 30%; CD4+ T cell IRR to Tet was 40%, and serum Ab IRR to LPV7 was 84%. These IRRs also trended higher for IFA-containing adjuvants (36% vs 18%, 48% vs 24%, and 97% vs 60%, respectively). CONCLUSIONS: The LPV7 vaccine is safe with each of seven adjuvant strategies and induced T cell responses to CD8 MEPs ex vivo in a subset of patients but did not enhance IRRs compared with prior vaccines using short peptides. Immunogenicity was supported more by IFA than by TLR agonists alone and may be enhanced by polyICLC plus IFA. TRIAL REGISTRATION NUMBER: NCT02126579.


Assuntos
Melanoma/tratamento farmacológico , Receptores Toll-Like/uso terapêutico , Vacinas de Subunidades Antigênicas/uso terapêutico , Feminino , Humanos , Masculino , Fatores de Risco , Vacinas de Subunidades Antigênicas/farmacologia
2.
J Immunother Cancer ; 9(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33479025

RESUMO

BACKGROUND: Peptide vaccines designed to stimulate melanoma-reactive CD4+ T cells can induce T cell and antibody (Ab) responses, associated with enhanced overall survival. We hypothesized that adding toll-like receptor 3 agonist polyICLC to an incomplete Freund's adjuvant (IFA) would be safe and would support strong, durable CD4+ T cell and Ab responses. We also hypothesized that oral low-dose metronomic cyclophosphamide (mCy) would be safe, would reduce circulating regulatory T cells (T-regs) and would further enhance immunogenicity. PARTICIPANTS AND METHODS: An adaptive design based on toxicity and durable CD4+ T cell immune response (dRsp) was used to assign participants with resected stage IIA-IV melanoma to one of four study regimens. The regimens included a vaccine comprising six melanoma peptides restricted by Class II MHC (6MHP) in an emulsion with IFA alone (Arm A), with IFA plus systemic mCy (Arm B), with IFA+ local polyICLC (Arm C), or with IFA+ polyICLC+ mCy (Arm D). Toxicities were recorded (CTCAE V.4.03). T cell responses were measured by interferon γ ELIspot assay ex vivo. Serum Ab responses to 6MHP were measured by ELISA. Circulating T-regs were assessed by flow cytometry. RESULTS: Forty-eight eligible participants were enrolled and treated. Early data on safety and dRsp favored enrollment on arm D. Total enrollment on Arms A-D were 3, 7, 6, and 32, respectively. Treatment-related dose-limiting toxicities (DLTs) were observed in 1/7 (14%) participants on arm B and 2/32 (6%) on arm D. None exceeded the 25% DLT threshold for early closure to enrollment for any arm. Strong durable T cell responses to 6MHP were detected ex vivo in 0%, 29%, 67%, and 47% of participants on arms A-D, respectively. IgG Ab responses were greatest for arms C and D. Circulating T-regs frequencies were not altered by mCy. CONCLUSIONS: 6MHP vaccines administered with IFA, polyICLC, and mCy were well tolerated. The dRsp rate for arm D of 47% (90% CI 32 to 63) exceeded the 18% (90% CI 11 to 26) rate previously observed with 6MHP in IFA alone. Vaccination with IFA+ polyICLC (arm C) also showed promise for enhancing T cell and Ab responses.


Assuntos
Carboximetilcelulose Sódica/análogos & derivados , Ciclofosfamida/administração & dosagem , Adjuvante de Freund/administração & dosagem , Lipídeos/administração & dosagem , Melanoma/tratamento farmacológico , Poli I-C/administração & dosagem , Polilisina/análogos & derivados , Vacinas de Subunidades Antigênicas/administração & dosagem , Administração Metronômica , Administração Oral , Anticorpos/sangue , Linfócitos T CD4-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Carboximetilcelulose Sódica/administração & dosagem , Carboximetilcelulose Sódica/efeitos adversos , Terapia Combinada , Ciclofosfamida/efeitos adversos , Feminino , Adjuvante de Freund/efeitos adversos , Humanos , Lipídeos/efeitos adversos , Masculino , Melanoma/imunologia , Melanoma/patologia , Estadiamento de Neoplasias , Poli I-C/efeitos adversos , Polilisina/administração & dosagem , Polilisina/efeitos adversos , Linfócitos T Reguladores/metabolismo , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/imunologia
3.
J Immunother Cancer ; 8(1)2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32385144

RESUMO

BACKGROUND: Phosphorylated peptides presented by MHC molecules represent a new class of neoantigens expressed on cancer cells and recognized by CD8 T-cells. These peptides are promising targets for cancer immunotherapy. Previous work identified an HLA-A*0201-restricted phosphopeptide from insulin receptor substrate 2 (pIRS2) as one such target. The purpose of this study was to characterize a second phosphopeptide, from breast cancer antiestrogen resistance 3 (BCAR3), and to evaluate safety and immunogenicity of a novel immunotherapic vaccine comprising either or both of these phosphorylated peptides. METHODS: Phosphorylated BCAR3 protein was evaluated in melanoma and breast cancer cell lines by Western blot, and recognition by T-cells specific for HLA-A*0201-restricted phosphorylated BCAR3 peptide (pBCAR3126-134) was determined by 51Cr release assay and intracellular cytokine staining. Human tumor explants were also evaluated by mass spectrometry for presentation of pIRS2 and pBCAR3 peptides. For the clinical trial, participants with resected stage IIA-IV melanoma were vaccinated 6 times over 12 weeks with one or both peptides in incomplete Freund's adjuvant and Hiltonol (poly-ICLC). Adverse events (AEs) were coded based on National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) V.4.03, with provision for early study termination if dose-limiting toxicity (DLT) rates exceeded 33%. The enrollment target was 12 participants evaluable for immune response to each peptide. T-cell responses were assessed by interferon-γ ELISpot assay. RESULTS: pBCAR3 peptides were immunogenic in vivo in mice, and in vitro in normal human donors, and T-cells specific for pBCAR3126-134 controlled outgrowth of a tumor xenograft. The pIRS21097-1105 peptide was identified by mass spectrometry from human hepatocellular carcinoma tumors. In the clinical trial, 15 participants were enrolled. All had grade 1 or 2 treatment-related AEs, but there were no grade 3-4 AEs, DLTs or deaths on study. T-cell responses were induced to the pIRS21097-1105 peptide in 5/12 patients (42%, 90% CI 18% to 68%) and to the pBCAR3126-134 peptide in 2/12 patients (17%, 90% CI 3% to 44%). CONCLUSION: This study supports the safety and immunogenicity of vaccines containing the cancer-associated phosphopeptides pBCAR3126-134 and pIRS21097-1105, and the data support continued development of immune therapy targeting phosphopeptides. Future studies will define ways to further enhance the magnitude and durability of phosphopeptide-specific immune responses. TRIAL REGISTRATION NUMBER: NCT01846143.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/efeitos adversos , Imunoterapia/efeitos adversos , Melanoma/terapia , Neoplasias Cutâneas/terapia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos de Neoplasias/genética , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/imunologia , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Humanos , Imunogenicidade da Vacina , Imunoterapia/métodos , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/imunologia , Masculino , Melanoma/imunologia , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fosfopeptídeos/genética , Fosfopeptídeos/imunologia , Projetos Piloto , Estudo de Prova de Conceito , Neoplasias Cutâneas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Immunother Cancer ; 5(1): 92, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29157306

RESUMO

BACKGROUND: Breast cancer remains a leading cause of cancer death worldwide. There is evidence that immunotherapy may play a role in the eradication of residual disease. Peptide vaccines for immunotherapy are capable of durable immune memory, but vaccines alone have shown sparse clinical activity against breast cancer to date. Toll-like receptor (TLR) agonists and helper peptides are excellent adjuvants for vaccine immunotherapy and they are examined in this human clinical trial. METHODS: A vaccine consisting of 9 MHC class I-restricted breast cancer-associated peptides (from MAGE-A1, -A3, and -A10, CEA, NY-ESO-1, and HER2 proteins) was combined with a TLR3 agonist, poly-ICLC, along with a helper peptide derived from tetanus toxoid. The vaccine was administered on days 1, 8, 15, 36, 57, 78. CD8+ T cell responses to the vaccine were assessed by both direct and stimulated interferon gamma ELIspot assays. RESULTS: Twelve patients with breast cancer were treated: five had estrogen receptor positive disease and five were HER2 amplified. There were no dose-limiting toxicities. Toxicities were limited to Grade 1 and Grade 2 and included mild injection site reactions and flu-like symptoms, which occurred in most patients. The most common toxicities were injection site reaction/induration and fatigue, which were experienced by 100% and 92% of participants, respectively. In the stimulated ELIspot assays, peptide-specific CD8+ T cell responses were detected in 4 of 11 evaluable patients. Two patients had borderline immune responses to the vaccine. The two peptides derived from CEA were immunogenic. No difference in immune response was evident between patients receiving endocrine therapy and those not receiving endocrine therapy during the vaccine series. CONCLUSIONS: Peptide vaccine administered in the adjuvant breast cancer setting was safe and feasible. The TLR3 adjuvant, poly-ICLC, plus helper peptide mixture provided modest immune stimulation. Further optimization is required for this multi-peptide vaccine/adjuvant combination. TRIAL REGISTRATION: ClinicalTrials.gov (posted 2/15/2012): NCT01532960. Registered 2/8/2012. https://clinicaltrials.gov/show/NCT01532960.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/análogos & derivados , Indutores de Interferon/uso terapêutico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Adjuvantes Imunológicos , Adulto , Vacinas Anticâncer/farmacologia , Carboximetilcelulose Sódica/farmacologia , Carboximetilcelulose Sódica/uso terapêutico , Feminino , Humanos , Imunoterapia , Indutores de Interferon/farmacologia , Pessoa de Meia-Idade , Projetos Piloto , Poli I-C/farmacologia , Polilisina/farmacologia , Polilisina/uso terapêutico
5.
PLoS Pathog ; 12(1): e1005382, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26761793

RESUMO

Generation of reactive oxygen species (ROS) during infection is an immediate host defense leading to microbial killing. APE1 is a multifunctional protein induced by ROS and after induction, protects against ROS-mediated DNA damage. Rac1 and NAPDH oxidase (Nox1) are important contributors of ROS generation following infection and associated with gastrointestinal epithelial injury. The purpose of this study was to determine if APE1 regulates the function of Rac1 and Nox1 during oxidative stress. Gastric or colonic epithelial cells (wild-type or with suppressed APE1) were infected with Helicobacter pylori or Salmonella enterica and assessed for Rac1 and NADPH oxidase-dependent superoxide production. Rac1 and APE1 interactions were measured by co-immunoprecipitation, confocal microscopy and proximity ligation assay (PLA) in cell lines or in biopsy specimens. Significantly greater levels of ROS were produced by APE1-deficient human gastric and colonic cell lines and primary gastric epithelial cells compared to control cells after infection with either gastric or enteric pathogens. H. pylori activated Rac1 and Nox1 in all cell types, but activation was higher in APE1 suppressed cells. APE1 overexpression decreased H. pylori-induced ROS generation, Rac1 activation, and Nox1 expression. We determined that the effects of APE1 were mediated through its N-terminal lysine residues interacting with Rac1, leading to inhibition of Nox1 expression and ROS generation. APE1 is a negative regulator of oxidative stress in the gastrointestinal epithelium during bacterial infection by modulating Rac1 and Nox1. Our results implicate APE1 in novel molecular interactions that regulate early stress responses elicited by microbial infections.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Mucosa Intestinal/imunologia , Infecções por Salmonella/imunologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Western Blotting , Linhagem Celular , Imunofluorescência , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/metabolismo , Humanos , Imunoprecipitação , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Microscopia Confocal , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Infecções por Salmonella/metabolismo , Proteínas rac1 de Ligação ao GTP/imunologia
6.
Mol Cell Biol ; 35(1): 303-17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368381

RESUMO

Breast cancer metastasis suppressor 1 (BRMS1) is downregulated in non-small cell lung cancer (NSCLC), and its reduction correlates with disease progression. Herein, we investigate the mechanisms through which loss of the BRMS1 gene contributes to epithelial-to-mesenchymal transition (EMT). Using a short hairpin RNA (shRNA) system, we show that loss of BRMS1 promotes basal and transforming growth factor beta-induced EMT in NSCLC cells. NSCLC cells expressing BRMS1 shRNAs (BRMS1 knockdown [BRMS1(KD)]) display mesenchymal characteristics, including enhanced cell migration and differential regulation of the EMT markers. Mesenchymal phenotypes observed in BRMS1(KD) cells are dependent on RelA/p65, the transcriptionally active subunit of nuclear factor kappa B (NF-κB). In addition, chromatin immunoprecipitation analysis demonstrates that loss of BRMS1 increases Twist1 promoter occupancy of RelA/p65 K310-a key histone modification associated with increased transcription. Knockdown of Twist1 results in reversal of BRMS1(KD)-mediated EMT phenotypic changes. Moreover, in our animal model, BRMS1(KD)/Twist1(KD) double knockdown cells were less efficient in establishing lung tumors than BRMS1(KD) cells. Collectively, this study demonstrates that loss of BRMS1 promotes malignant phenotypes that are dependent on NF-κB-dependent regulation of Twist1. These observations offer fresh insight into the mechanisms through which BRMS1 regulates the development of metastases in NSCLC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Transição Epitelial-Mesenquimal , Genes Supressores de Tumor , Histonas/metabolismo , Humanos , Metástase Linfática , Camundongos , Camundongos Nus , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Nucleares/genética , Fenótipo , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/genética , Fator de Crescimento Transformador beta/metabolismo , Proteína 1 Relacionada a Twist/genética , Cicatrização
7.
PLoS One ; 9(4): e95869, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24763730

RESUMO

Expression of the breast cancer metastasis suppressor 1 (BRMS1) protein is dramatically reduced in non-small cell lung cancer (NSCLC) cells and in primary human tumors. Although BRMS1 is a known suppressor of metastasis, the mechanisms through which BRMS1 functions to regulate cell migration and invasion in response to specific NSCLC driver mutations are poorly understood. To experimentally address this, we utilized immortalized human bronchial epithelial cells in which p53 was knocked down in the presence of oncogenic K-RasV12 (HBEC3-p53KD-K-RasV12). These genetic alterations are commonly found in NSCLC and are associated with a poor prognosis. To determine the importance of BRMS1 for cytoskeletal function, cell migration and invasion in our model system we stably knocked down BRMS1. Here, we report that loss of BRMS1 in HBEC3-p53KD-K-RasV12 cells results in a dramatic increase in cell migration and invasion compared to controls that expressed BRMS1. Moreover, the loss of BRMS1 resulted in additional morphological changes including F-actin re-distribution, paxillin accumulation at the leading edge of the lamellapodium, and cellular shape changes resembling mesenchymal phenotypes. Importantly, re-expression of BRMS1 restores, in part, cell migration and invasion; however it does not fully reestablish the epithelial phenotype. These finding suggests that loss of BRMS1 results in a permanent, largely irreversible, mesenchymal phenotype associated with increased cell migration and invasion. Collectively, in NSCLC cells without p53 and expression of oncogenic K-Ras our study identifies BRMS1 as a key regulator required to maintain a cellular morphology and cytoskeletal architecture consistent with an epithelial phenotype.


Assuntos
Células Epiteliais Alveolares/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Proteínas ras/genética , Citoesqueleto de Actina/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Técnicas de Silenciamento de Genes , Humanos , Camundongos Nus , Mutação , Paxilina/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Proteínas Repressoras , Proteínas ras/metabolismo
8.
Cancer Res ; 73(4): 1308-17, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23269275

RESUMO

The mechanisms through which the metastasis suppressor gene BRMS1 functions are poorly understood. Herein, we report the identification of a previously undescribed E3 ligase function of BRMS1 on the histone acetyltransferase p300. BRMS1 induces polyubiquitination of p300, resulting in its proteasome-mediated degradation. We identify BRMS1 as the first eukaryote structural mimic of the bacterial IpaH E3 ligase family and establish that the evolutionarily conserved CXD motif located in BRMS1 is responsible for its E3 ligase function. Mutation of this E3 ligase motif not only abolishes BRMS1-induced p300 polyubiquitination and degradation, but importantly, dramatically reduces the metastasis suppressor function of BRMS1 in both in vitro and in vivo models of lung cancer metastasis.


Assuntos
Proteína p300 Associada a E1A/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proteína p300 Associada a E1A/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Mutação , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Interferência de RNA , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transplante Heterólogo , Carga Tumoral/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/genética
9.
Dig Dis ; 29(2): 249-54, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21734392

RESUMO

There is growing evidence that many aspects of our lifestyle and the environment we now live in contribute to the development of disease. The luminal digestive tract is a clear target of the influence of dietary components, alcohol, microbial organisms, and other ingested materials. External factors including obesity, lack of physical exercise, and tobacco consumption also impact diseases of the luminal gastrointestinal (GI) tract. A growing understanding of the microbiome which forms an integral part of the human organism indicates that this is another important external force that impacts human health and disease. The luminal GI tract conditions that arise, at least in part, from these external factors range from malignancies (squamous cell esophageal cancer, Barrett's esophagus and associated esophageal adenocarcinoma, gastric cancer, and colorectal cancer), idiopathic inflammatory disorders such as inflammatory bowel diseases, and post-infectious syndromes including post-infectious irritable bowel syndrome, post-infectious dyspepsia and other functional GI disorders. Of particular interest, given their increase in prevalence in much of the world, are immune-mediated conditions in which food antigens are the driving force behind disease development. These entities include celiac disease, eosinophilic esophagitis, and food allergies. Celiac disease is a prime example of a condition mediated by dietary factors whose pathogenesis has only recently been determined, providing opportunities for developing treatment options beyond the gluten-free diet. While a genetic basis for this disease clearly exists, it is believed that environmental factors such as an increase in gluten in the human diet account for its rising prevalence, now roughly 1% of genetically susceptible populations in all continents. Proposed therapeutic strategies span from preventing disease by modulating the time of gluten introduction in infants, to reducing exposure to gluten by developing strains of wheat with lower levels of gluten, degrading ingested gluten peptides within the intestinal lumen via endopeptidases or modulating uptake of these peptides across intestinal tight junctions. Other novel treatments in development focus on interfering with the immune events that lead to disease once gluten accesses the lamina propria including altering the immune milieu from a Th1-predominant response via hookworm infection, inhibiting tissue transglutaminase, and blocking antigen presentation and/or T-cell responses to gluten peptides. While new treatment options for celiac disease reflect the complex interaction of diet, genetic factors and the host immune response, the implications for treatment of many conditions of the large and small intestine that arise from environmental and lifestyle are as basic as ensuring adequate nutrition, regular exercise and cessation of tobacco use. Much more needs to be learned about the microbiome, dietary and other factors and their interaction with the human host in order to develop potential new treatment strategies for diseases that result from the environment and lifestyle.


Assuntos
Meio Ambiente , Enteropatias/epidemiologia , Enteropatias/terapia , Intestino Grosso/patologia , Intestino Delgado/patologia , Estilo de Vida , Hipersensibilidade Alimentar/complicações , Humanos , Enteropatias/complicações
10.
Cell Signal ; 23(12): 1972-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21787862

RESUMO

Streptolysin O (SLO) is a protein cytotoxin derived from Group A beta-hemolytic streptococci that associates with membranes and permeabilizes cells. Oxidation inactivates SLO, eliminating the characteristic hemolytic and cytotoxic activities. However, oxidized SLO produces beneficial therapeutic effects in vivo on scleroderma, scar formation and wound healing. Here we report that oxidized SLO also significantly inhibited invasion by human metastatic breast cancer MDA-MB-231 cells through Matrigel in an in vitro model of metastatic disease. This dose-dependent response corresponded to selective SLO activation of epidermal growth factor receptor (EGFR) ErbB1. SLO and EGF were equally selective in activation of EGFR, but EGF elicited larger relative increases in phosphorylation at various sites, especially pronounced for Tyr845. Addition of SLO did not affect either ERK1/2 or Akt kinases and altered the expression of only 10 of 84 metastasis-related genes in MDA-MB-231 cells. Neither SLO nor EGF promoted growth of several human breast cancer cell lines. Knockdown of EGFR by siRNA ablated the inhibitory effect of SLO on cancer cell invasion, showing SLO selectively activated ErbB1 kinase to reduce invasion without increasing cell growth. The results suggest SLO might have promise as a new therapy to inhibit metastasis.


Assuntos
Neoplasias da Mama/patologia , Colágeno/química , Receptores ErbB/agonistas , Laminina/química , Proteoglicanas/química , Estreptolisinas/farmacologia , Proteínas de Bactérias/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Combinação de Medicamentos , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Genes Neoplásicos , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação , Interferência de RNA
11.
Am J Physiol Gastrointest Liver Physiol ; 299(5): G1177-86, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20829524

RESUMO

Hypoxia-inducible factor 1 (HIF1) consists of a hypoxia-inducible α subunit and a constitutively expressed ß subunit. Reactive oxygen species (ROS) induced by Helicobacter pylori stabilize HIF1α in the human gastric epithelium in normoxia. HIF1α plays crucial role in carcinogenesis and has been associated with malignant progression of gastric cancer. Several genes contain functional hypoxia-response elements (HREs) in their promoters including Bcl2 family member, Mcl1. Cellular ratios of antiapoptotic oncogenic protein, Mcl1, and tumor suppressor proapoptotic protein, Noxa, determine cell fate by regulating normal cellular growth, cell death and oncogenic processes. The aim of the present study was to examine the mechanism of HIF1α induction in the H. pylori-infected gastric epithelium to better understand disease pathogenesis by H. pylori relevant to gastric carcinogenesis. Our data showed that the dose-dependent increase in HIF1α in H. pylori-infected gastric epithelia is mediated by induction of a ROS-inducible protein, apurinic/apyrimidinic endonuclease 1 (APE1), and an enhanced interaction of APE1 with the transcriptional coactivator p300. Surprisingly, with accumulation of HIF1α, further transcriptional activation of mcl1 was not observed. We identified a HIF-binding site (HBS) in the hif1α promoter and showed that increased HIF1α expression, whether H. pylori-induced or hypoxia-mimetic agent, CoCl(2)-induced, resulted in enhanced HIF1α binding to its own promoter. This resulted in a transcriptionally inactive hif1α promoter since hif1α HBS lacks HIF ancillary sequence (HAS) required for HIF1 transcriptional activity. We conclude that enhanced binding of "nonfunctional" HIF1α to hif1α promoter and limiting availability of p300 in the cell serves as checkpoints for uncontrolled HIF1α activity.


Assuntos
Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Análise de Variância , Western Blotting , Células Epiteliais/microbiologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estômago/microbiologia , Transfecção
12.
Mol Cell Proteomics ; 9(12): 2853-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20798394

RESUMO

Tensin1 is the archetype of a family of focal adhesion proteins. Tensin1 has a phosphotyrosine binding domain that binds the cytoplasmic tail of ß-integrin, a Src homology 2 domain that binds focal adhesion kinase, p130Cas, and the RhoGAP called deleted in liver cancer-1, a phosphatase and tensin homology domain that binds protein phosphatase-1α and other regions that bind F-actin. The association between tensin1 and these partners affects cell polarization, migration, and invasion. In this study we analyzed the phosphorylation of human S-tag-tensin1 expressed in HEK293 cells by mass spectrometry. Peptides covering >90% of the sequence initially revealed 50 phosphorylated serine/phosphorylated threonine (pSer/pThr) but no phosphorylated tyrosine (pTyr) sites. Addition of peroxyvanadate to cells to inhibit protein tyrosine phosphatases exposed 10 pTyr sites and addition of calyculin A to cells to inhibit protein phosphatases type 1 and 2A gave a total of 62 pSer/pThr sites. We also characterized two sites modified by O-linked N-acetylglucosamine. Tensin1 F302A, which does not bind protein phosphatase-1, showed > twofold enhanced phosphorylation of seven sites. The majority of pSer/pThr have adjacent proline (Pro) residues and we show endogenous p38 mitogen activated protein kinase (MAPK) associated with and phosphorylated tensin1 in an in vitro kinase assay. Recombinant p38α MAPK also phosphorylated S-tag-tensin1, resulting in decreased binding with deleted in liver cancer-1. Activation of p38 MAPK in cells by sorbitol-induced hyperosmotic stress increased phosphorylation of S-tag-tensin1, which reduced binding to deleted in liver cancer-1 and increased binding to endogenous pTyr proteins, including p130Cas and focal adhesion kinase. These data demonstrate that tensin1 is extensively phosphorylated on Ser/Thr residues in cells and phosphorylation by p38 MAPK regulates the specificity of the tensin1 Src homology 2 domain for binding to different proteins. Tensin1 provides a hub for connecting signaling pathways involving p38 MAP kinase, tyrosine kinases and RhoGTPases.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acilação , Sequência de Aminoácidos , Linhagem Celular , Cromatografia de Afinidade , Cromatografia Líquida , Humanos , Proteínas dos Microfilamentos/química , Dados de Sequência Molecular , Fosforilação , Espectrometria de Massas em Tandem , Tensinas , Domínios de Homologia de src
13.
J Biol Chem ; 284(50): 34713-22, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19826001

RESUMO

Tensin is a family of multidomain scaffold proteins that bind the cytoplasmic tail of beta-integrins and localize to adhesions that anchor stress fibers in cells. Tensin expression is suppressed in cancer, especially metastatic cancer. The N-terminal domain of tensin1 associates with protein phosphatase-1alpha (PP1alpha) and mediates PP1alpha localization to adhesions. Here, we show F302A mutation in a KVXF motif of tensin1 abrogates binding to PP1alpha. The SH2 domain in tensin family member c-ten requires R474 to bind a RhoGAP called DLC-1 (deleted in liver cancer). We mutated the corresponding residue in tensin1, R1488A, and showed this reduces association with DLC-1. Unexpectedly, tensin1 F302A also had reduced association with DLC-1. Expression of tensin1 F302A or R1488A showed similar dominant phenotypes, with reduced cell polarization, lowered MLC20 phosphorylation and reduced levels of RhoA(GTP) compared with cells expressing tensin1 WT. However, migration and invasion of metastatic MDA MB 231 breast cancer cells were differentially affected by tensin1 mutated at F302A or R1488A. Cancer cells stably expressing F302A tensin1 showed increased migration and invasion compared with cells stably expressing either R1488A tensin1 or WT tensin1. This suggests that PP1alpha bound to tensin1 has additional effects in reducing migration and invasion that are not mediated through DLC-1. Our results show the importance of PP1alpha binding to tensin1 for the regulation of cell polarization, migration, and invasion.


Assuntos
Movimento Celular/fisiologia , Polaridade Celular , Proteínas dos Microfilamentos/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Adesão Celular/fisiologia , Linhagem Celular , Forma Celular , Ativação Enzimática , Proteínas Ativadoras de GTPase , Humanos , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Invasividade Neoplásica/fisiopatologia , Neoplasias/metabolismo , Neoplasias/patologia , Mutação Puntual , Proteína Fosfatase 1/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Tensinas , Proteínas Supressoras de Tumor/genética , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Apoptosis ; 12(9): 1721-31, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17520193

RESUMO

Non-ionizing radiation produced by nanosecond pulsed electric fields (nsPEFs) is an alternative to ionizing radiation for cancer treatment. NsPEFs are high power, low energy (non-thermal) pulses that, unlike plasma membrane electroporation, modulate intracellular structures and functions. To determine functions for p53 in nsPEF-induced apoptosis, HCT116p53(+/+) and HCT116p53(-/-) colon carcinoma cells were exposed to multiple pulses of 60 kV/cm with either 60 ns or 300 ns durations and analyzed for apoptotic markers. Several apoptosis markers were observed including cell shrinkage and increased percentages of cells positive for cytochrome c, active caspases, fragmented DNA, and Bax, but not Bcl-2. Unlike nsPEF-induced apoptosis in Jurkat cells (Beebe et al. 2003a) active caspases were observed before increases in cytochrome c, which occurred in the presence and absence of Bax. Cell shrinkage occurred only in cells with increased levels of Bax or cytochrome c. NsPEFs induced apoptosis equally in HCT116p53(+/+) and HCT116p53(-/-) cells. These results demonstrate that non-ionizing radiation produced by nsPEFs can act as a non-ligand agonist with therapeutic potential to induce apoptosis utilizing mitochondrial-independent mechanisms in HCT116 cells that lead to caspase activation and cell death in the presence or absence of p-53 and Bax.


Assuntos
Apoptose/efeitos da radiação , Campos Eletromagnéticos , Proteína Supressora de Tumor p53/fisiologia , Caspases/metabolismo , Citocromos c/biossíntese , Fragmentação do DNA/efeitos da radiação , Indução Enzimática , Células HCT116 , Humanos , Radiação não Ionizante , Proteína X Associada a bcl-2/metabolismo
15.
DNA Cell Biol ; 26(3): 160-71, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17417944

RESUMO

Nanosecond pulsed electric fields (nsPEFs) are a type of nonthermal, nonionizing radiation that exhibit intense electric fields with high power, but low energy. NsPEFs extend conventional electroporation (EP) to affect intracellular structures and functions and depending on the intensity, can induce lethal and nonlethal cell signaling. In this study, HCT116 human colon carcinoma cells were synchronized to the S-phase or remained unsynchronized, exposed to electric fields of 60 kV/cm with either 60-ns or 300-ns durations, and analyzed for apoptosis and proliferative markers. Several nsPEF structural and functional targets were identified. Unlike unsynchronized cells, S-phase cells under limiting conditions exhibited greater membrane integrity and caspase activation and maintained cytoskeletal structure. Regardless of synchronization, cells exposed to nsPEFs under these conditions primarily survived, but exhibited some turnover and delayed proliferation in cell populations, as well as reversible increases in phosphatidylserine externalization, membrane integrity, and nuclei size. These results show that nsPEFs can act as a nonligand agonist to modulate plasma membrane (PM) and intracellular structures and functions, as well as differentially affect cells in the S-phase, but without effect on cell survival. Furthermore, nsPEF effects on the nucleus and cytoskeleton may provide synergistic therapeutic actions with other agents, such as ionizing radiation or chemotherapeutics that affect these same structures.


Assuntos
Proliferação de Células/efeitos da radiação , Eletricidade , Fase S/efeitos da radiação , Actinas/metabolismo , Actinas/efeitos da radiação , Caspases/metabolismo , Diferenciação Celular , Membrana Celular/efeitos da radiação , Núcleo Celular/efeitos da radiação , Sobrevivência Celular , Citoplasma/efeitos da radiação , Humanos , Radiação não Ionizante , Fase S/efeitos dos fármacos , Timidina/farmacologia , Células Tumorais Cultivadas
16.
DNA Cell Biol ; 24(5): 283-91, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15869405

RESUMO

Nanosecond pulsed electric fields (nsPEFs) are ultrashort pulses with high electric field intensity (kV/cm) and high power (megawatts), but low energy density (mJ/cc). To determine roles for p53 in response to nsPEFs, HCT116 cells (p53+/+ and p53-/-) were exposed to nsPEF and analyzed for membrane integrity, phosphatidylserine externalization, caspase activation, and cell survival. Decreasing plasma membrane effects were observed in both HCT116p53+/+ and p53-/- cells with decreasing pulse durations and/or decreasing electric fields. However, addition of ethidium homodimer-1 and Annexin-V-FITC post-pulse demonstrated greater fluorescence in p53-/- versus p53+/+ cells, suggesting a postpulse p53-dependent biological effect at the plasma membrane. Caspase activity was significantly higher than nonpulsed cells only in the p53-/- cells. HCT116 cells exhibited greater survival in response to nsPEFs than HL-60 and Jurkat cells, but survival was more evident for HCT116p53+/+ cells than for HCT116p53-/- cells. These results indicate that nsPEF effects on HCT116 cells include (1) apparent direct electric field effects, (2) biological effects that are p53-dependent and p53-independent, (3) actions on mechanisms that originate at the plasma membranes and at intracellular structures, and (4) an apparent p53 protective effect. NsPEF applications provide a means to explore intracellular structures and functions that can reveal mechanisms in health and disease.


Assuntos
Caspases/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efeitos da radiação , Campos Eletromagnéticos , Etídio/análogos & derivados , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/efeitos da radiação , Anexina A5 , Inibidores de Caspase , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Ativação Enzimática , Etídio/farmacologia , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Proteínas de Fluorescência Verde/metabolismo , Células HCT116 , Células HL-60 , Humanos , Hidrazinas , Substâncias Intercalantes/farmacologia , Células Jurkat , Microscopia Confocal , Fosfatidilserinas/análise , Fatores de Tempo
17.
Immunogenetics ; 55(2): 87-94, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12698287

RESUMO

To detect minor barriers to histocompatibility that might be encoded on the X chromosome in mice, we grafted reciprocal sets of (C57BL/6xBALB/c)F1, (C57BL/6xDBA/2)F1, and (BALB/cxDBA/2)F1 mice with tail skin from the respective paternal inbred strain. Our histogenic analysis suggests that, compared with the C57BL/6 mouse strain, the BALB/c strain generates X-linked antigen loss. In contrast, we detected no X-linked histogenic differences between strains C57BL/6 and DBA/2, or DBA/2 and BALB/c. To localize this X-linked barrier to histocompatibility, we produced a panel of 25 [(BALB/cxC57BL/6)F1xC57BL/6]N2 males that were grafted with C57BL/6 skin to determine which carried the BALB/c-derived component(s) necessary for graft rejection. DNA marker analysis showed one region of overlapping BALB/c-derived X-chromosomal segments among the graft rejecters, suggesting that this antigen-loss haplotype ( H-hix(c), for histoincompatibility on the X chromosome, c haplotype) may be restricted within the DXMit55 to the Xq telomere interval (which excludes only the centromeric tip of the X). Further backcrossing of H-hix(c) to C57BL/6 resulted in fewer rejecter mice than expected by the N4 generation, suggesting that a second, unlinked locus is also involved in this X-linked alloantigenicity. The vigorous rejection of male (C57BL/6xBALB)F1 and female (B6.C- H2(d)xC57BL/6)F1 skin by (BALB/cxC57BL/6)F1 males, as well as the assessment of markers on Chromosome 17 among N2 and N4 graft-recipient males, suggests that this second locus is H2, and that H-hix(b)-encoded alloantigens require both H2(b) and H2(d)-encoded presentation molecules for efficient graft rejection.


Assuntos
Camundongos Endogâmicos BALB C/imunologia , Camundongos Endogâmicos C57BL/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Animais , Cruzamentos Genéticos , Feminino , Rejeição de Enxerto/imunologia , Masculino , Camundongos , Antígenos de Histocompatibilidade Menor/genética , Transplante de Pele/imunologia , Fatores de Tempo , Tolerância ao Transplante/imunologia , Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...