Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Radiat Res ; 189(5): 490-496, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29528769

RESUMO

Amifostine is a potent antioxidant that protects against ionizing radiation effects. In this study, we evaluated the effect of Amifostine administered before total-body irradiation (TBI), at a drug dose that protects against TBI lethality, for potential protection against radiation-induced late effects such as a shortened lifespan and cancer. Three groups of mice were studied: 0 Gy control; 10.8 Gy TBI with Amifostine pretreatment; and 5.4 Gy TBI alone. Animals were monitored for their entire lifespan. The median survival times for mice receiving 0, 5.4 or 10.8 Gy TBI were 706, 460 and 491 days, respectively. Median survival of both irradiated groups was significantly shorter compared to nonirradiated mice ( P < 0.0001). Cancer incidence (hematopoietic and solid tumors) was similar between the irradiated groups and was significantly greater than for the 0 Gy controls. The ratio of hematopoietic-to-solid tumors differed among the groups, with the 5.4 Gy group having a higher incidence of hematopoietic neoplasms compared to the 10.8 Gy/Amifostine group (1.8-fold). Solid tumor incidence was greater in the 10.8 Gy/Amifostine group (1.6-fold). There are few mouse lifespan studies for agents that protect against radiation-induced lethality. Mice treated with 10.8 Gy/Amifostine yielded a lower incidence of hematopoietic neoplasms and higher incidence of solid neoplasms. In conclusion, mice protected from lethal TBI have a shortened lifespan, due in large part to cancer induction after exposure compared to nonexposed controls. Amifostine treatment did protect against radiation-induced hematopoietic tumors, while protection against solid neoplasms was significant but incomplete.


Assuntos
Amifostina/farmacologia , Neoplasias Induzidas por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Irradiação Corporal Total/efeitos adversos , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Camundongos , Neoplasias Induzidas por Radiação/etiologia
2.
Inflamm Bowel Dis ; 23(11): 1983-1995, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29019857

RESUMO

BACKGROUND: If treatment with intravenous steroids fail, inflammatory bowel disease patients with acute severe colitis face systemic anti-tumor necrosis factor biologic rescue therapy or colectomy. Interleukin (IL)-27 is a cytokine with an immunosuppressive role in adaptive immune responses. However, the IL-27 receptor complex is also expressed on innate immune cells, and there is evidence that IL-27 can impact the function of innate cell subsets, although this particular functionality in vivo is not understood. Our aim was to define the efficacy of IL-27 in acute severe colitis and characterize novel IL-27-driven mechanisms of immunosuppression in the colonic mucosa. METHODS: We assessed oral delivery of Lactococcus lactis expressing an IL-27 hyperkine on the innate immune response in vivo in a genetically intact, noninfective, acute murine colitis model induced by intrarectal instillation of 2,4,6-trinitrobenzenesulfonic acid in SJL/J mice. RESULTS: IL-27 attenuates acute severe colitis through the reduction of colonic mucosal neutrophil infiltrate associated with a decreased CXC chemokine gradient. This suppression was T cell independent and IL-10 dependent, initially featuring enhanced mucosal IL-10. IL-27 was associated with a reduction in colonic proinflammatory cytokines and induced a multifocal, strong, positive nuclear expression of phosphorylated STAT-1 in mucosal epithelial cells. CONCLUSION: We have defined novel mechanisms of IL-27 immunosuppression toward colonic innate immune responses in vivo. Mucosal delivery of IL-27 has translational potential as a novel therapeutic for inflammatory bowel disease, and it is a future mucosal directed rescue therapy in acute severe inflammatory bowel disease.


Assuntos
Colite/tratamento farmacológico , Colo/imunologia , Imunidade Inata , Interleucina-10/metabolismo , Interleucina-27/farmacologia , Mucosa Intestinal/metabolismo , Doença Aguda , Animais , Colite/induzido quimicamente , Colo/fisiopatologia , Modelos Animais de Doenças , Inflamação/patologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/patologia , Interleucina-27/imunologia , Mucosa Intestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Linfócitos T/metabolismo , Ácido Trinitrobenzenossulfônico/administração & dosagem
3.
Oncotarget ; 8(19): 30621-30643, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28430642

RESUMO

Effective drug development to combat metastatic disease in breast cancer would be aided by the availability of well-characterized preclinical animal models that (a) metastasize with high efficiency, (b) metastasize in a reasonable time-frame, (c) have an intact immune system, and (d) capture some of the heterogeneity of the human disease. To address these issues, we have assembled a panel of twelve mouse mammary cancer cell lines that can metastasize efficiently on implantation into syngeneic immunocompetent hosts. Genomic characterization shows that more than half of the 30 most commonly mutated genes in human breast cancer are represented within the panel. Transcriptomically, most of the models fall into the luminal A or B intrinsic molecular subtypes, despite the predominance of an aggressive, poorly-differentiated or spindled histopathology in all models. Patterns of immune cell infiltration, proliferation rates, apoptosis and angiogenesis differed significantly among models. Inherent within-model variability of the metastatic phenotype mandates large cohort sizes for intervention studies but may also capture some relevant non-genetic sources of variability. The varied molecular and phenotypic characteristics of this expanded panel of models should aid in model selection for development of antimetastatic therapies in vivo, and serve as a useful platform for predictive biomarker identification.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias Mamárias Experimentais , Aloenxertos , Animais , Biomarcadores Tumorais , Biópsia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Genômica/métodos , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Terapia de Alvo Molecular , Metástase Neoplásica , Polimorfismo de Nucleotídeo Único
4.
PLoS One ; 11(4): e0153270, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27077656

RESUMO

Laser capture microdissection (LCM) of tissue is an established tool in medical research for collection of distinguished cell populations under direct microscopic visualization for molecular analysis. LCM samples have been successfully analyzed in a number of genomic and proteomic downstream molecular applications. However, LCM sample collection and preparation procedure has to be adapted to each downstream analysis platform. In this present manuscript we describe in detail the adaptation of LCM methodology for the collection and preparation of fresh frozen samples for NanoString analysis based on a study of a model of mouse mammary gland carcinoma and its lung metastasis. Our adaptation of LCM sample preparation and workflow to the requirements of the NanoString platform allowed acquiring samples with high RNA quality. The NanoString analysis of such samples provided sensitive detection of genes of interest and their associated molecular pathways. NanoString is a reliable gene expression analysis platform that can be effectively coupled with LCM.


Assuntos
Microdissecção e Captura a Laser/métodos , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/patologia , Nanotecnologia/métodos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Secções Congeladas/métodos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Mamárias Animais/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Estabilidade de RNA , Reprodutibilidade dos Testes
5.
Cancer Res ; 76(6): 1569-77, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26880804

RESUMO

Nonlethal exposure to ionizing radiation (IR) is a public concern due to its known carcinogenic effects. Although latency periods for IR-induced neoplasms are relatively long, the ability to detect cancer as early as possible is highly advantageous for effective therapeutic intervention. Therefore, we hypothesized that metabolites in the urine from mice exposed to total body radiation (TBI) would predict for the presence of cancer before a palpable mass was detected. In this study, we exposed mice to 0 or 5.4 Gy TBI, collected urine samples periodically over 1 year, and assayed urine metabolites by using mass spectrometry. Longitudinal data analysis within the first year post-TBI revealed that cancers, including hematopoietic, solid, and benign neoplasms, could be distinguished by unique urinary signatures as early as 3 months post-TBI. Furthermore, a distinction among different types of malignancies could be clearly delineated as early as 3 months post-TBI for hematopoietic neoplasms, 6 months for solid neoplasms, and by 1 year for benign neoplasms. Moreover, the feature profile for radiation-exposed mice 6 months post-TBI was found to be similar to nonirradiated control mice at 18 months, suggesting that TBI accelerates aging. These results demonstrate that urine feature profiles following TBI can identify cancers in mice prior to macroscopic detection, with important implications for the early diagnosis and treatment.


Assuntos
Neoplasias Induzidas por Radiação/metabolismo , Animais , Relação Dose-Resposta à Radiação , Feminino , Espectrometria de Massas/métodos , Metabolômica/métodos , Camundongos , Camundongos Endogâmicos C3H , Radiação Ionizante , Irradiação Corporal Total/métodos
6.
J Autoimmun ; 53: 33-45, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24583068

RESUMO

We generated a mouse model with a 162 nt AU-rich element (ARE) region deletion in the 3' untranslated region (3'UTR) of the interferon-gamma (IFN-γ) gene that results in chronic circulating serum IFN-γ levels. Mice homozygous for the ARE deletion (ARE-Del) (-/-) present both serologic and cellular abnormalities typical of patients with systemic lupus erythematosus (SLE). ARE-Del(-/-) mice display increased numbers of pDCs in bone marrow and spleen. Addition of IFN-γ to Flt3-ligand (Flt3L) treated in vitro bone marrow cultures results in a 2-fold increase in pDCs with concurrent increases in IRF8 expression. Marginal zone B (MZB) cells and marginal zone macrophages (MZMs) are absent in ARE-Del(-/-) mice. ARE-Del(+/-) mice retain both MZB cells and MZMs and develop no or mild autoimmunity. However, low dose clodronate treatment in ARE-Del(+/-) mice specifically eliminates MZMs and promotes anti-DNA antibody development and glomerulonephritis. Our findings demonstrate the consequences of a chronic IFN-γ milieu on B220(+) cell types and in particular the impact of MZB cell loss on MZM function in autoimmunity. Furthermore, similarities between disease states in ARE-Del(-/-) mice and SLE patients suggest that IFN-γ may not only be a product of SLE but may be critical for disease onset and progression.


Assuntos
Elementos Ricos em Adenilato e Uridilato/genética , Sequência de Bases , Interferon gama , Nefrite Lúpica/imunologia , Deleção de Sequência , Animais , Anticorpos Antinucleares/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Interferon gama/genética , Interferon gama/imunologia , Nefrite Lúpica/genética , Macrófagos/imunologia , Macrófagos/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout
7.
Gastroenterology ; 146(1): 210-221.e13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24120477

RESUMO

BACKGROUND & AIMS: Treatment of inflammatory bowel disease would benefit from specific targeting of therapeutics to the intestine. We developed a strategy for localized delivery of the immunosuppressive cytokine interleukin (IL)-27, which is synthesized actively in situ by the food-grade bacterium Lactococcus lactis (LL-IL-27), and tested its ability to reduce colitis in mice. METHODS: The 2 genes encoding mouse IL-27 were synthesized with optimal codon use for L lactis and joined with a linker; a signal sequence was added to allow for product secretion. The construct was introduced into L lactis. Colitis was induced via transfer of CD4(+)CD45RB(hi) T cells into Rag(-/-) mice to induce colitis; 7.5 weeks later, LL-IL-27 was administered to mice via gavage. Intestinal tissues were collected and analyzed. RESULTS: LL-IL-27 administration protected mice from T-cell transfer-induced enterocolitis and death. LL-IL-27 reduced disease activity scores, pathology features of large and small bowel, and levels of inflammatory cytokines in colonic tissue. LL-IL-27 also reduced the numbers of CD4(+) and IL-17(+) T cells in gut-associated lymphoid tissue. The effects of LL-IL-27 required production of IL-10 by the transferred T cells. LL-IL-27 was more effective than either LL-IL-10 or systemic administration of recombinant IL-27 in reducing colitis in mice. LL-IL-27 also reduced colitis in mice after administration of dextran sodium sulfate. CONCLUSIONS: LL-IL-27 reduces colitis in mice by increasing the production of IL-10. Mucosal delivery of LL-IL-27 could be a more effective and safer therapy for inflammatory bowel disease.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Enterocolite/imunologia , Fatores Imunológicos/administração & dosagem , Doenças Inflamatórias Intestinais , Interleucina-10/imunologia , Interleucinas/administração & dosagem , Mucosa Intestinal/imunologia , Lactococcus lactis , Administração Oral , Animais , Modelos Animais de Doenças , Fatores Imunológicos/farmacologia , Interleucinas/imunologia , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Linfócitos T , Transformação Bacteriana
8.
Pigment Cell Melanoma Res ; 27(1): 37-47, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24128326

RESUMO

Melanoma represents a significant malignancy in humans and dogs. Different from genetically engineered models, sporadic canine melanocytic neoplasms share several characteristics with human disease that could make dogs a more relevant preclinical model. Canine melanomas rarely arise in sun-exposed sites. Most occur in the oral cavity, with a subset having intra-epithelial malignant melanocytes mimicking the in situ component of human mucosal melanoma. The spectrum of canine melanocytic neoplasia includes benign lesions with some analogy to nevi, as well as invasive primary melanoma, and widespread metastasis. Growing evidence of distinct subtypes in humans, differing in somatic and predisposing germ-line genetic alterations, cell of origin, epidemiology, relationship to ultraviolet radiation and progression from benign to malignant tumors, may also exist in dogs. Canine and human mucosal melanomas appear to harbor BRAF, NRAS, and c-kit mutations uncommonly, compared with human cutaneous melanomas, although both species share AKT and MAPK signaling activation. We conclude that there is significant overlap in the clinical and histopathological features of canine and human mucosal melanomas. This represents opportunity to explore canine oral cavity melanoma as a preclinical model.


Assuntos
Doenças do Cão , Melanoma , Neoplasias Bucais , Neoplasias Cutâneas , Animais , Modelos Animais de Doenças , Doenças do Cão/genética , Doenças do Cão/metabolismo , Doenças do Cão/patologia , Cães , Humanos , Sistema de Sinalização das MAP Quinases/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Melanoma/veterinária , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Neoplasias Bucais/veterinária , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/veterinária
9.
Proc Natl Acad Sci U S A ; 110(33): 13534-9, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23904478

RESUMO

Increased serum levels of IL-15 are reported in type 1 diabetes (T1D). Here we report elevated serum soluble IL-15Rα levels in human T1D. To investigate the role of IL-15/IL-15Rα in the pathogenesis of T1D, we generated double transgenic mice with pancreatic ß-cell expression of IL-15 and IL-15Rα. The mice developed hyperglycemia, marked mononuclear cell infiltration, ß-cell destruction, and anti-insulin autoantibodies that mimic early human T1D. The diabetes in this model was reversed by inhibiting IL-15 signaling with anti-IL2/IL15Rß (anti-CD122), which blocks IL-15 transpresentation. Furthermore, the diabetes could be reversed by administration of the Janus kinase 2/3 inhibitor tofacitinib, which blocks IL-15 signaling. In an alternative diabetes model, nonobese diabetic mice, IL15/IL-15Rα expression was increased in islet cells in the prediabetic stage, and inhibition of IL-15 signaling with anti-CD122 at the prediabetic stage delayed diabetes development. In support of the view that these observations reflect the conditions in humans, we demonstrated pancreatic islet expression of both IL-15 and IL-15Rα in human T1D. Taken together our data suggest that disordered IL-15 and IL-15Rα may be involved in T1D pathogenesis and the IL-15/IL15Rα system and its signaling pathway may be rational therapeutic targets for early T1D.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Modelos Animais de Doenças , Células Secretoras de Insulina/metabolismo , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Interleucina-15/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos , Interleucina-15/antagonistas & inibidores , Interleucina-15/sangue , Subunidade alfa de Receptor de Interleucina-15/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Piperidinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia
10.
Free Radic Biol Med ; 65: 497-508, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23851018

RESUMO

Reactive oxygen species generated by NADPH oxidase 5 (Nox5) have been implicated in physiological and pathophysiological signaling pathways, including cancer development and progression. However, because immunological tools are lacking, knowledge of the role of Nox5 in tumor biology has been limited; the expression of Nox5 protein across tumors and normal tissues is essentially unknown. Here, we report the characterization and use of a mouse monoclonal antibody against a recombinant Nox5 protein (bp 600-746) for expression profiling of Nox5 in human tumors by tissue microarray analysis. Using our novel antibody, we also report the detection of endogenous Nox5 protein in human UACC-257 melanoma cells. Immunofluorescence, confocal microscopy, and immunohistochemical techniques were employed to demonstrate Nox5 localization throughout UACC-257 cells, with perinuclear enhancement. Tissue microarray analysis revealed, for the first time, substantial Nox5 overexpression in several human cancers, including those of prostate, breast, colon, lung, brain, and ovary, as well as in malignant melanoma and non-Hodgkin lymphoma; expression in most nonmalignant tissues was negative to weak. This validated mouse monoclonal antibody will promote further exploration of the functional significance of Nox5 in human pathophysiology, including tumor cell growth and proliferation.


Assuntos
Anticorpos Monoclonais , Biomarcadores Tumorais/análise , Proteínas de Membrana/biossíntese , NADPH Oxidases/biossíntese , Neoplasias/enzimologia , Animais , Western Blotting , Linhagem Celular Tumoral , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/análise , Camundongos , Microscopia Confocal , NADPH Oxidase 5 , NADPH Oxidases/análise , Reação em Cadeia da Polimerase em Tempo Real , Análise Serial de Tecidos
11.
PLoS One ; 8(2): e57389, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23460847

RESUMO

Changes in dietary selenium and selenoprotein status may influence both anti- and pro-cancer pathways, making the outcome of interventions different from one study to another. To characterize such outcomes in a defined setting, we undertook a controlled hepatocarcinogenesis study involving varying levels of dietary selenium and altered selenoprotein status using mice carrying a mutant (A37G) selenocysteine tRNA transgene (Trsp(tG37) ) and/or a cancer driver TGFα transgene. The use of Trsp(tG37) altered selenoprotein expression in a selenoprotein and tissue specific manner and, at sufficient dietary selenium levels, separate the effect of diet and selenoprotein status. Mice were maintained on diets deficient in selenium (0.02 ppm selenium) or supplemented with 0.1, 0.4 or 2.25 ppm selenium or 30 ppm triphenylselenonium chloride (TPSC), a non-metabolized selenium compound. Trsp(tG37) transgenic and TGFα/Trsp(tG37) bi-transgenic mice subjected to selenium-deficient or TPSC diets developed a neurological phenotype associated with early morbidity and mortality prior to hepatocarcinoma development. Pathology analyses revealed widespread disseminated pyogranulomatous inflammation. Pyogranulomas occurred in liver, lungs, heart, spleen, small and large intestine, and mesenteric lymph nodes in these transgenic and bi-transgenic mice. The incidence of liver tumors was significantly increased in mice carrying the TGFα transgene, while dietary selenium and selenoprotein status did not affect tumor number and multiplicity. However, adenoma and carcinoma size and area were smaller in TGFα transgenic mice that were fed 0.4 and 2.25 versus 0.1 ppm of selenium. Thus, selenium and selenoprotein deficiencies led to widespread pyogranuloma formation, while high selenium levels inhibited the size of TGFα-induced liver tumors.


Assuntos
Suplementos Nutricionais , Granuloma/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Selênio/deficiência , Selênio/uso terapêutico , Selenoproteínas/deficiência , Animais , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Granuloma/sangue , Isótopos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/sangue , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , RNA de Transferência Aminoácido-Específico/metabolismo , Selênio/sangue , Selenoproteínas/metabolismo , Fator de Crescimento Transformador alfa
12.
Infect Immun ; 81(2): 585-97, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23230288

RESUMO

We employed Francisella tularensis live vaccine strain (LVS) to study mechanisms of protective immunity against intracellular pathogens and, specifically, to understand protective correlates. One potential molecular correlate identified previously was interleukin-6 (IL-6), a cytokine with pleotropic roles in immunity, including influences on T and B cell functions. Given its role as an immune modulator and the correlation with successful anti-LVS vaccination, we examined the role IL-6 plays in the host response to LVS. IL-6-deficient (IL-6 knockout [KO]) mice infected with LVS intradermally or intranasally or anti-IL-6-treated mice, showed greatly reduced 50% lethal doses compared to wild-type (WT) mice. Increased susceptibility was not due to altered splenic immune cell populations during infection or decreased serum antibody production, as IL-6 KO mice had similar compositions of each compared to WT mice. Although LVS-infected IL-6 KO mice produced much less serum amyloid A and haptoglobin (two acute-phase proteins) than WT mice, there were no other obvious pathophysiological differences between LVS-infected WT and IL-6 KO mice. IL-6 KO or WT mice that survived primary LVS infection also survived a high-dose LVS secondary challenge. Using an in vitro overlay assay that measured T cell activation, cytokine production, and abilities of primed splenocytes to control intracellular LVS growth, we found that IL-6 KO total splenocytes or purified T cells were slightly defective in controlling intracellular LVS growth but were equivalent in cytokine production. Taken together, IL-6 is an integral part of a successful immune response to primary LVS infection, but its exact role in precipitating adaptive immunity remains elusive.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Interleucina-6/imunologia , Tularemia/imunologia , Animais , Vacinas Bacterianas/metabolismo , Vacinas Bacterianas/farmacologia , Francisella tularensis/metabolismo , Haptoglobinas/imunologia , Haptoglobinas/metabolismo , Interleucina-6/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Amiloide A Sérica/imunologia , Proteína Amiloide A Sérica/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Tularemia/metabolismo , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/metabolismo , Vacinas Atenuadas/farmacologia
13.
Cancer Res ; 72(18): 4846-55, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22805306

RESUMO

There is significant interest in the development of agents that can ameliorate radiation damage after exposure to radiation has occurred. Here we report that chronic supplementation of the antioxidant Tempol in the diet of mice can reduce body weight without toxicity, decrease cancer, and extend survival when administered after nonlethal total body radiation (TBI). These effects were apparent in two different strains of mice (C3H, CBA) exposed to TBI (3 Gy). Notably, delaying administration of the Tempol diet one month after TBI could also enhance survival. Tempol reduced the incidence of hematopoietic neoplasms (lymphomas) in both strains, whereas both the onset and incidence of nonhematopoietic neoplasms were reduced in CBA mice. These results encourage further study of Tempol as a chemopreventive, to reduce the incidence of radiation-induced second malignancies after a course of definitive radiation therapy. Tempol may also find applications to reduce the risk of cancers in populations exposed to nonlethal radiation due to nuclear accidents or terrorist attacks.


Assuntos
Antioxidantes/farmacologia , Óxidos N-Cíclicos/farmacologia , Neoplasias Induzidas por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Irradiação Corporal Total/efeitos adversos , Animais , Feminino , Camundongos , Marcadores de Spin
14.
Nat Commun ; 3: 884, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22673911

RESUMO

Malignant melanoma of the skin (CMM) is associated with ultraviolet radiation exposure, but the mechanisms and even the wavelengths responsible are unclear. Here we use a mammalian model to investigate melanoma formed in response to precise spectrally defined ultraviolet wavelengths and biologically relevant doses. We show that melanoma induction by ultraviolet A (320-400 nm) requires the presence of melanin pigment and is associated with oxidative DNA damage within melanocytes. In contrast, ultraviolet B radiation (280-320 nm) initiates melanoma in a pigment-independent manner associated with direct ultraviolet B DNA damage. Thus, we identified two ultraviolet wavelength-dependent pathways for the induction of CMM and describe an unexpected and significant role for melanin within the melanocyte in melanomagenesis.


Assuntos
Melaninas/metabolismo , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanoma/etiologia , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Cromatografia Líquida de Alta Pressão , Dano ao DNA/efeitos dos fármacos , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Imuno-Histoquímica , Masculino , Melaninas/genética , Camundongos , Microscopia Confocal , Neoplasias Induzidas por Radiação/genética , Neoplasias Induzidas por Radiação/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
PLoS One ; 7(12): e52655, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285131

RESUMO

BACKGROUND: Despite high cure rates for pediatric B-lineage acute lymphoblastic leukemia (B-ALL), short-term and long-term toxicities and chemoresistance are shortcomings of standard chemotherapy. Immunotherapy and chemoimmunotherapy based on monoclonal antibodies (mAbs) that target cell surface antigens with restricted expression in pediatric B-ALL may offer the potential to reduce toxicities and prevent or overcome chemoresistance. The receptor tyrosine kinase ROR1 has emerged as a candidate for mAb targeting in select B-cell malignancies. METHODOLOGY AND PRINCIPAL FINDINGS: Using flow cytometry, Western blotting, immunohistochemistry, and confocal immunofluorescence microscopy, we analyzed the cell surface expression of ROR1 across major pediatric ALL subtypes represented by 14 cell lines and 56 primary blasts at diagnosis or relapse as well as in normal adult and pediatric tissues. Cell surface ROR1 expression was found in 45% of pediatric ALL patients, all of which were B-ALL, and was not limited to any particular genotype. All cell lines and primary blasts with E2A-PBX1 translocation and a portion of patients with other high risk genotypes, such as MLL rearrangement, expressed cell surface ROR1. Importantly, cell surface ROR1 expression was found in many of the pediatric B-ALL patients with multiply relapsed and refractory disease and normal karyotype or low risk cytogenetics, such as hyperdiploidy. Notably, cell surface ROR1 was virtually absent in normal adult and pediatric tissues. CONCLUSIONS AND SIGNIFICANCE: Collectively, this study suggests that ROR1 merits preclinical and clinical investigations as a novel target for mAb-based therapies in pediatric B-ALL. We propose cell surface expression of ROR1 detected by flow cytometry as primary inclusion criterion for pediatric B-ALL patients in future clinical trials of ROR1-targeted therapies.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Adolescente , Adulto , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Lactente , Masculino , Terapia de Alvo Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/antagonistas & inibidores , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Adulto Jovem
17.
Int J Cancer ; 126(2): 459-68, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19585575

RESUMO

Cancer cells undergo significant changes in carbohydrate expression, and these alterations can be useful as biomarkers and therapeutic targets. In this study, we investigated the expression of carbohydrate antigens containing a terminal GalNAcalpha1-3Gal or GalNAcalpha1-6Gal on human cervix and cervical carcinoma. Monoclonal antibodies to each of these carbohydrates were generated by immunizing rabbits with the corresponding antigen conjugated to KLH followed by hybridoma production. Antibodies were screened and evaluated using a combination of carbohydrate microarray profiling, ELISA, dot blot and immunohistochemical staining to verify specificity. Antibody 132-3 was found to selectively recognize GalNAcalpha1-3Gal with little cross-reactivity to other structurally similar antigens such as GalNAcalpha1-6Gal, blood group A, Forssman antigen and the Tn antigen on both solution assays and human tissue. Although GalNAcalpha1-6Gal expression was not detected, GalNAcalpha1-3Gal expression was found on 55% of squamous cell carcinomas. Expression in normal tissue was observed but was restricted to the suprabasal epithelial layer. Importantly, we found expression of the antigen on cervical cancer had a statistically significant correlation with the 5-year survival rate of the patients (48 vs. 85% for antigen negative vs. positive, p = 0.017). Expression of GalNAcalpha1-3Gal did not correlate with other clinical factors including tumor stage, size and lymph node metastasis, indicating the antigen is a new, independent biomarker for the prognosis of cervical cancer.


Assuntos
Biomarcadores Tumorais/análise , Dissacarídeos/análise , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/metabolismo , Adulto , Anticorpos Monoclonais/imunologia , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/metabolismo , Colo do Útero/química , Colo do Útero/patologia , Distribuição de Qui-Quadrado , Dissacarídeos/química , Dissacarídeos/imunologia , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metástase Linfática , Pessoa de Meia-Idade , Estrutura Molecular , Prognóstico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
18.
PLoS One ; 4(11): e7637, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19907640

RESUMO

Interleukin-7 (IL-7) is required for lymphocyte development and homeostasis although the actual sites of IL-7 production have never been clearly identified. We produced a bacterial artificial chromosome (BAC) transgenic mouse expressing ECFP in the Il7 locus. The construct lacked a signal peptide and ECFP (enhanced cyan fluorescent protein) accumulated inside IL-7-producing stromal cells in thoracic thymus, cervical thymus and bone marrow. In thymus, an extensive reticular network of IL-7-containing processes extended from cortical and medullary epithelial cells, closely contacting thymocytes. Central memory CD8 T cells, which require IL-7 and home to bone marrow, physically associated with IL-7-producing cells as we demonstrate by intravital imaging.


Assuntos
Interleucina-7/genética , Linfócitos/citologia , Animais , Medula Óssea/metabolismo , Linfócitos T CD8-Positivos/citologia , Separação Celular , Cromossomos Artificiais Bacterianos , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Interleucina-7/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Recombinação Genética , Timo/metabolismo
19.
Mol Cancer Ther ; 8(7): 1867-77, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19584228

RESUMO

Inhibition of hypoxia inducible factor-1 (HIF-1) is an attractive therapeutic strategy to target the tumor microenvironment. However, HIF-1 inhibitors may have limited activity as single agents and combination therapies may be required. We tested the hypothesis that HIF-1 inhibition in a hypoxic-stressed tumor microenvironment, which could be generated by administration of antiangiogenic agents, may result in a more pronounced therapeutic effect. The activity of bevacizumab, either alone or in combination with the HIF-1alpha inhibitor topotecan, was evaluated in U251-HRE xenografts. Tumor tissue was collected at the end of treatment and changes in tumor oxygenation, angiogenesis, proliferation, apoptosis, HIF-1alpha levels, HIF-1 target genes, and DNA damage were evaluated. Bevacizumab decreased microvessel-density and increased intratumor-hypoxia, but did not induce apoptosis. Moreover, bevacizumab alone caused a significant increase of HIF-1-dependent gene expression in tumor tissue. Addition of a low dose of daily topotecan to bevacizumab significantly inhibited tumor growth, relative to mice treated with topotecan or bevacizumab alone (P < 0.01). The addition of topotecan to bevacizumab was also associated with profound inhibition of HIF-1 transcriptional activity, significant inhibition of proliferation, and induction of apoptosis. Importantly, DNA damage induced by topotecan alone was not augmented by addition of bevacizumab, suggesting that increased cytotoxic activity did not account for the increased antitumor effects observed. These results strongly suggest that combination of anti-vascular endothelial growth factor antibodies with HIF-1 inhibitors is an attractive therapeutic strategy targeting in the hypoxic tumor microenvironment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Bevacizumab , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Neoplasias Encefálicas/parasitologia , Neoplasias Encefálicas/patologia , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Glioma/irrigação sanguínea , Glioma/patologia , Humanos , Técnicas Imunoenzimáticas , Luciferases/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica/prevenção & controle , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Topotecan/administração & dosagem , Células Tumorais Cultivadas
20.
Mol Cancer Ther ; 8(4): 971-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19372570

RESUMO

The tumor-associated Tn antigen has been investigated extensively as a biomarker and therapeutic target. Cancer vaccines containing the Tn antigen as a single tumor antigen or as a component of a polyvalent vaccine have progressed into phase I and II clinical trials. One major focus of Tn-based vaccines is the treatment of prostate cancer patients. Although expression of the antigen on prostate tumors is a critical prerequisite, previous reports investigating Tn expression in prostate tumors have produced conflicting results. Using a combination of immunohistochemistry and carbohydrate microarray profiling, we show that only 4% to 26% of prostate tumors express the Tn antigen. Based on our results, the majority of prostate cancer patients do not express the appropriate antigen. Therefore, efforts to preselect the subset of prostate cancer patients with Tn-positive tumors or apply Tn vaccines to other cancers with higher rates of antigen expression could significantly improve clinical response rates. Because conflicting information on carbohydrate expression is a general problem for the field, the approach described in this article of analyzing antigen expression with multiple antibodies and using carbohydrate microarray profiles to interpret the results will be useful for the development of other carbohydrate-based cancer vaccines and diagnostics.


Assuntos
Antígenos Glicosídicos Associados a Tumores/metabolismo , Biomarcadores Tumorais/metabolismo , Vacinas Anticâncer/uso terapêutico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/terapia , Idoso , Animais , Antígenos Glicosídicos Associados a Tumores/imunologia , Biomarcadores Tumorais/imunologia , Carboidratos/imunologia , Carcinoma de Células de Transição/imunologia , Carcinoma de Células de Transição/metabolismo , Carcinoma de Células de Transição/terapia , Ensaios Clínicos como Assunto , Humanos , Hiperplasia/imunologia , Hiperplasia/metabolismo , Hiperplasia/terapia , Masculino , Neoplasias da Próstata/metabolismo , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...