Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS One ; 15(6): e0233846, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32502202

RESUMO

Growth Differentiation Factor-15 (GDF15) is a divergent TGF-beta superfamily cytokine that is overexpressed by most cancers and is induced by anticancer therapy. Transgenic and induced animal models suggest that it protects from cancer development but the mechanisms are uncertain. We investigated the role of immunity in GDF15 induced reduction in prostate cancer (PCa) growth. The C57BL/6 transgenic TRAMP prostate cancer prone mice were bred with mice that were immunodeficient and/or systemically overexpressed GDF15. We developed a novel orthotopic TRAMP PCa model in which primary TRAMP tumor cells were implanted into prostates of mice to reduce the study time. These mice were administered recombinant mouse GDF15, antibody to CD8, PD1 or their respective controls. We found that GDF15 induced protection from tumor growth was reversed by lack of adaptive immunity. Flow cytometric evaluation of lymphocytes within these orthotopic tumors showed that GDF15 overexpression was associated with increased CD8 T cell numbers and an increased number and proportion of recently activated CD8+CD11c+ T cells and a reduced proportion of "exhausted" CD8+PD1+ T cells. Further, depletion of CD8 T cells in tumor bearing mice abolished the GDF15 induced protection from tumor growth. Infusion of GDF15 into mice bearing orthotopic TRAMP tumor, substantially reduced tumor growth that was further reduced by concurrent PD1 antibody administration. GDF15 overexpression or recombinant protein protects from TRAMP tumor growth by modulating CD8 T cell mediated antitumor immunity and augments the positive effects of anti-PD1 blockers.


Assuntos
Antineoplásicos/uso terapêutico , Fator 15 de Diferenciação de Crescimento/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Feminino , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Experimentais
2.
Int J Obes (Lond) ; 43(12): 2370-2380, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31152154

RESUMO

BACKGROUND: Elevated circulating levels of the divergent transforming growth factor-beta (TGFb) family cytokine, growth differentiation factor 15 (GDF15), acting through its CNS receptor, glial-derived neurotrophic factor receptor alpha-like (GFRAL), can cause anorexia and weight loss leading to anorexia/cachexia syndrome of cancer and other diseases. Preclinical studies suggest that administration of drugs based on recombinant GDF15 might be used to treat severe obesity. However, the role of the GDF15-GFRAL pathway in the physiological regulation of body weight and metabolism is unclear. The critical site of action of GFRAL in the CNS has also not been proven beyond doubt. To investigate these two aspects, we have inhibited the actions of GDF15 in mice started on high-fat diet (HFD). METHODS: The actions of GDF15 were inhibited using two methods: (1) Groups of 8 mice under HFD had their endogenous GDF15 neutralised by monoclonal antibody treatment, (2) Groups of 15 mice received AAV-shRNA to knockdown GFRAL at its hypothesised major sites of action, the hindbrain area postrema (AP) and the nucleus of the solitary tract (NTS). Metabolic measurements were determined during both experiments. CONCLUSIONS: Treating mice with monoclonal antibody to GDF15 shortly after commencing HFD results in more rapid gain of body weight, adiposity and hepatic lipid deposition than the control groups. This is accompanied by reduced glucose and insulin tolerance and greater expression of pro-inflammatory cytokines in adipose tissue. Localised AP and NTS shRNA-GFRAL knockdown in mice commencing HFD similarly caused an increase in body weight and adiposity. This effect was in proportion to the effectiveness of GFRAL knockdown, indicated by quantitative analysis of hindbrain GFRAL staining. We conclude that the GDF15-GFRAL axis plays an important role in resistance to obesity in HFD-fed mice and that the major site of action of GDF15 in the CNS is GFRAL-expressing neurons in the AP and NTS.


Assuntos
Adiposidade , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Fator 15 de Diferenciação de Crescimento , Rombencéfalo , Adiposidade/genética , Adiposidade/fisiologia , Animais , Área Postrema/citologia , Área Postrema/metabolismo , Área Postrema/fisiologia , Peso Corporal/fisiologia , Dieta Hiperlipídica , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Obesidade/metabolismo , Rombencéfalo/citologia , Rombencéfalo/metabolismo , Rombencéfalo/fisiologia , Núcleo Solitário/citologia , Núcleo Solitário/metabolismo , Núcleo Solitário/fisiologia
3.
Cell Metab ; 28(3): 353-368, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30184485

RESUMO

MIC-1/GDF15 is a stress response cytokine and a distant member of the transforming growth factor beta (TGFb) superfamily, with no close relatives. It acts via a recently identified receptor called glial-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL), which is a distant orphan member of the GDNF receptor family that signals through the tyrosine kinase receptor Ret. MIC-1/GDF15 expression and serum levels rise in response to many stimuli that initiate cell stress and as part of a wide variety of disease processes, most prominently cancer and cardiovascular disease. The best documented actions of MIC-1/GDF15 are on regulation of energy homeostasis. When MIC-1/GDF15 serum levels are substantially elevated in diseases like cancer, it subverts a physiological pathway of appetite regulation to induce an anorexia/cachexia syndrome initiated by its actions on hindbrain neurons. These effects make it a potential target for the treatment of both obesity and anorexia/cachexia syndromes, disorders lacking any highly effective, readily accessible therapies.


Assuntos
Caquexia/metabolismo , Metabolismo Energético/fisiologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Obesidade/metabolismo , Animais , Anorexia/metabolismo , Doenças Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Homeostase , Humanos , Inflamação/metabolismo , Camundongos , Doenças Mitocondriais/metabolismo , Neoplasias/metabolismo , Ratos
4.
Biol Open ; 5(5): 620-30, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-27113959

RESUMO

Intracellular chloride channel protein 1 (CLIC1) participates in inflammatory processes by regulating macrophage phagosomal functions such as pH and proteolysis. Here, we sought to determine if CLIC1 can regulate adaptive immunity by actions on dendritic cells (DCs), the key professional antigen presenting cells. To do this, we first generated bone marrow-derived DCs (BMDCs) from germline CLIC1 gene-deleted (CLIC1(-/-)) and wild-type (CLIC1(+/+)) mice, then studied them in vitro and in vivo We found phagocytosis triggered cytoplasmic CLIC1 translocation to the phagosomal membrane where it regulated phagosomal pH and proteolysis. Phagosomes from CLIC1(-/-) BMDCs displayed impaired acidification and proteolysis, which could be reproduced if CLIC1(+/+), but not CLIC1(-/-) cells, were treated with IAA94, a CLIC family ion channel blocker. CLIC1(-/-) BMDC displayed reduced in vitro antigen processing and presentation of full-length myelin oligodendrocyte glycoprotein (MOG) and reduced MOG-induced experimental autoimmune encephalomyelitis. These data suggest that CLIC1 regulates DC phagosomal pH to ensure optimal processing of antigen for presentation to antigen-specific T-cells. Further, they indicate that CLIC1 is a novel therapeutic target to help reduce the adaptive immune response in autoimmune diseases.

5.
PLoS One ; 10(7): e0133362, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26207898

RESUMO

The TGF-b superfamily cytokine MIC-1/GDF15 circulates in the blood of healthy humans. Its levels rise substantially in cancer and other diseases and this may sometimes lead to development of an anorexia/cachexia syndrome. This is mediated by a direct action of MIC-1/GDF15 on feeding centres in the hypothalamus and brainstem. More recent studies in germline gene deleted mice also suggest that this cytokine may play a role in physiological regulation of energy homeostasis. To further characterize the role of MIC-1/GDF15 in physiological regulation of energy homeostasis in man, we have examined diurnal and food associated variation in serum levels and whether variation in circulating levels relate to BMI in human monozygotic twin pairs. We found that the within twin pair differences in serum MIC-1/GDF15 levels were significantly correlated with within twin pair differences in BMI, suggesting a role for MIC-1/GDF15 in the regulation of energy balance in man. MIC-1/GDF15 serum levels altered slightly in response to a meal, but comparison with variation its serum levels over a 24 hour period suggested that these changes are likely to be due to bimodal diurnal variation which can alter serum MIC-1/GDF15 levels by about plus or minus 10% from the mesor. The lack of a rapid and substantial postprandial increase in MIC-1/GDF15 serum levels suggests that MIC1/GDF15 is unlikely to act as a satiety factor. Taken together, our findings suggest that MIC-1/GDF15 may be a physiological regulator of energy homeostasis in man, most probably due to actions on long-term regulation of energy homeostasis.


Assuntos
Índice de Massa Corporal , Ritmo Circadiano/fisiologia , Fator 15 de Diferenciação de Crescimento/sangue , Período Pós-Prandial/fisiologia , Saciação/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colecistocinina/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Saciação/efeitos dos fármacos , Gêmeos , Adulto Jovem
6.
PLoS One ; 10(2): e0115189, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25695521

RESUMO

The divergent TGF-ß superfamily member, macrophage inhibitory cytokine-1 (MIC-1/GDF15), is overexpressed by most cancers, including prostate cancer (PCa). Whilst its circulating levels are linked to cancer outcome, the role MIC-1/GDF15 plays in cancer development and progression is incompletely understood. To investigate its effect on PCa development and spread, we have used TRAMP prostate cancer prone mice bearing a germline deletion of MIC-1/GDF15 (TRAMPMIC-/-). On average TRAMPMIC-/- mice died about 5 weeks earlier and had larger prostatic tumors compared with TRAMP mice that were wild type for MIC-1/GDF15 (TRAMPMIC+/+). Additionally, at the time of death or ethical end point, even when adjusted for lifespan, there were no significant differences in the number of mice with metastases between the TRAMPMIC+/+ and TRAMPMIC-/- groups. However, consistent with our previous data, more than twice as many TRAMP mice overexpressing MIC-1/GDF15 (TRAMPfmsmic-1) had metastases than TRAMPMIC+/+ mice (p<0.0001). We conclude that germ line gene deletion of MIC-1/GDF15 leads to increased local tumor growth resulting in decreased survival consistent with an overall protective role for MIC-1/GDF15 in early primary tumor development. However, in advancing disease, as we have previously noted, MIC-1/GDF15 overexpression may promote local invasion and metastatic spread.


Assuntos
Fator 15 de Diferenciação de Crescimento/deficiência , Fator 15 de Diferenciação de Crescimento/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Animais , Deleção de Genes , Fator 15 de Diferenciação de Crescimento/genética , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/genética
7.
PLoS One ; 9(6): e100370, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24971956

RESUMO

Macrophage inhibitory cytokine-1 (MIC-1/GDF15) modulates food intake and body weight under physiological and pathological conditions by acting on the hypothalamus and brainstem. When overexpressed in disease, such as in advanced cancer, elevated serum MIC-1/GDF15 levels lead to an anorexia/cachexia syndrome. To gain a better understanding of its actions in the brainstem we studied MIC-1/GDF15 induced neuronal activation identified by induction of Fos protein. Intraperitoneal injection of human MIC-1/GDF15 in mice activated brainstem neurons in the area postrema (AP) and the medial (m) portion of the nucleus of the solitary tract (NTS), which did not stain with tyrosine hydroxylase (TH). To determine the importance of these brainstem nuclei in the anorexigenic effect of MIC-1/GDF15, we ablated the AP alone or the AP and the NTS. The latter combined lesion completely reversed the anorexigenic effects of MIC-1/GDF15. Altogether, this study identified neurons in the AP and/or NTS, as being critical for the regulation of food intake and body weight by MIC-1/GDF15.


Assuntos
Depressores do Apetite/farmacologia , Área Postrema/efeitos dos fármacos , Área Postrema/fisiologia , Fator 15 de Diferenciação de Crescimento/farmacologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/fisiologia , Animais , Anorexia/induzido quimicamente , Depressores do Apetite/administração & dosagem , Fator 15 de Diferenciação de Crescimento/administração & dosagem , Infusões Intraventriculares , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Redução de Peso/efeitos dos fármacos
8.
PLoS One ; 8(2): e55174, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23468844

RESUMO

The TGF-b superfamily cytokine MIC-1/GDF15 circulates in all humans and when overproduced in cancer leads to anorexia/cachexia, by direct action on brain feeding centres. In these studies we have examined the role of physiologically relevant levels of MIC-1/GDF15 in the regulation of appetite, body weight and basal metabolic rate. MIC-1/GDF15 gene knockout mice (MIC-1(-/-)) weighed more and had increased adiposity, which was associated with increased spontaneous food intake. Female MIC-1(-/-) mice exhibited some additional alterations in reduced basal energy expenditure and physical activity, possibly owing to the associated decrease in total lean mass. Further, infusion of human recombinant MIC-1/GDF15 sufficient to raise serum levels in MIC-1(-/-) mice to within the normal human range reduced body weight and food intake. Taken together, our findings suggest that MIC-1/GDF15 is involved in the physiological regulation of appetite and energy storage.


Assuntos
Apetite/genética , Peso Corporal/genética , Fator 15 de Diferenciação de Crescimento/genética , Tecido Adiposo/crescimento & desenvolvimento , Animais , Apetite/fisiologia , Peso Corporal/fisiologia , Ingestão de Alimentos , Metabolismo Energético/genética , Feminino , Genótipo , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Fatores Sexuais , Transdução de Sinais , Aumento de Peso/genética
9.
J Cachexia Sarcopenia Muscle ; 3(4): 239-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22936174

RESUMO

Anorexia/cachexia is a common and currently mostly untreatable complication of advanced cancer. It is also a feature of a number of chronic diseases and can also occur as part of the normal ageing process. Over recent years, two different, but sometimes overlapping, processes have been identified to mediate anorexia/cachexia: those that act primarily on muscle reducing its mass and function, and processes that decrease nutrition leading to loss of both fat and muscle. In the case of at least some cancers, the latter process is sometimes driven by marked overexpression of macrophage inhibitory cytokine-1/growth differentiation factor 15 (MIC-1/GDF15). MIC-1/GDF15 is a transforming growth factor beta (TGF-ß) family cytokine that is found in the serum of all normal individuals at an average concentration of about 0.6 ng/ml. Its increased expression in both cancers and other diseases can result in 10-100-fold or more elevation of its serum levels. In experimental animals, serum MIC-1/GDF15 levels at the lower end of this range induce anorexia by direct actions of the circulating cytokine on feeding centres in the brain. Mice with tumours overexpressing MIC-1/GDF15 display decreased food intake, loss of lean and fat mass and cachexia. That this process also mediates anorexia/cachexia in humans is suggested by the fact that there is a direct correlation between the degree of serum MIC-1/GDF15 elevation and the amount of cancer-related weight loss, the first such relationship demonstrated. Further, in experimental animals, weight loss can be reversed by neutralisation of tumour-produced MIC-1/GDF15 with a specific monoclonal antibody, suggesting the possibility of effective therapy of patients with the devastating complication of anorexia/cachexia.

10.
J Cell Sci ; 125(Pt 22): 5479-88, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22956539

RESUMO

Intracellular chloride channel protein 1 (CLIC1) is a 241 amino acid protein of the glutathione S transferase fold family with redox- and pH-dependent membrane association and chloride ion channel activity. Whilst CLIC proteins are evolutionarily conserved in Metazoa, indicating an important role, little is known about their biology. CLIC1 was first cloned on the basis of increased expression in activated macrophages. We therefore examined its subcellular localisation in murine peritoneal macrophages by immunofluorescence confocal microscopy. In resting cells, CLIC1 is observed in punctate cytoplasmic structures that do not colocalise with markers for endosomes or secretory vesicles. However, when these macrophages phagocytose serum-opsonised zymosan, CLIC1 translocates onto the phagosomal membrane. Macrophages from CLIC1(-/-) mice display a defect in phagosome acidification as determined by imaging live cells phagocytosing zymosan tagged with the pH-sensitive fluorophore Oregon Green. This altered phagosomal acidification was not accompanied by a detectable impairment in phagosomal-lysosomal fusion. However, consistent with a defect in acidification, CLIC1(-/-) macrophages also displayed impaired phagosomal proteolytic capacity and reduced reactive oxygen species production. Further, CLIC1(-/-) mice were protected from development of serum transfer induced K/BxN arthritis. These data all point to an important role for CLIC1 in regulating macrophage function through its ion channel activity and suggest it is a suitable target for the development of anti-inflammatory drugs.


Assuntos
Ácidos/metabolismo , Canais de Cloreto/metabolismo , Macrófagos Peritoneais/metabolismo , Fagossomos/metabolismo , Animais , Artrite/metabolismo , Artrite/patologia , Proteínas do Citoesqueleto/metabolismo , Glicolatos/farmacologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/enzimologia , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , NADPH Oxidases/metabolismo , Fagossomos/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína RAC2 de Ligação ao GTP
11.
PLoS One ; 7(8): e43833, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952779

RESUMO

Macrophage inhibitory cytokine-1 (MIC-1/GDF15), a divergent member of the TGF-ß superfamily, is over-expressed by many common cancers including those of the prostate (PCa) and its expression is linked to cancer outcome. We have evaluated the effect of MIC-1/GDF15 overexpression on PCa development and spread in the TRAMP transgenic model of spontaneous prostate cancer. TRAMP mice were crossed with MIC-1/GDF15 overexpressing mice (MIC-1(fms)) to produce syngeneic TRAMP(fmsmic-1) mice. Survival rate, prostate tumor size, histopathological grades and extent of distant organ metastases were compared. Metastasis of TC1-T5, an androgen independent TRAMP cell line that lacks MIC-1/GDF15 expression, was compared by injecting intravenously into MIC-1(fms) and syngeneic C57BL/6 mice. Whilst TRAMP(fmsmic-1) survived on average 7.4 weeks longer, had significantly smaller genitourinary (GU) tumors and lower PCa histopathological grades than TRAMP mice, more of these mice developed distant organ metastases. Additionally, a higher number of TC1-T5 lung tumor colonies were observed in MIC-1(fms) mice than syngeneic WT C57BL/6 mice. Our studies strongly suggest that MIC-1/GDF15 has complex actions on tumor behavior: it limits local tumor growth but may with advancing disease, promote metastases. As MIC-1/GDF15 is induced by all cancer treatments and metastasis is the major cause of cancer treatment failure and cancer deaths, these results, if applicable to humans, may have a direct impact on patient care.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , Neoplasias da Próstata/patologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Suscetibilidade a Doenças , Feminino , Fator 15 de Diferenciação de Crescimento/genética , Masculino , Camundongos , Camundongos Transgênicos , Gradação de Tumores , Metástase Neoplásica , Neoplasias da Próstata/genética , Análise de Sobrevida
12.
Growth Factors ; 29(5): 187-95, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21831009

RESUMO

Macrophage inhibitory cytokine-1 (MIC-1/GDF15) is associated with cardiovascular disease, inflammation, body weight regulation and cancer. Its serum levels facilitate the diagnosis and prognosis of cancer and vascular disease. Furthermore, its serum levels are a powerful predictor of all-cause mortality, suggesting a fundamental role in biological processes associated with ageing. In cancer, the data available suggest that MIC-1/GDF15 is antitumorigenic, but this may not always be the case as disease progresses. Cancer promoting effects of MIC-1/GDF15 may be due, in part, to effects on antitumour immunity. This is suggested by the anti-inflammatory and immunosuppressive properties of MIC-1/GDF15 in animal models of atherosclerosis and rheumatoid arthritis. Furthermore, in late-stage cancer, large amounts of MIC-1/GDF15 in the circulation suppress appetite and mediate cancer anorexia/cachexia, which can be reversed by monoclonal antibodies in animals. Available data suggest MIC-1/GDF15 may be an important molecule mediating the interplay between cancer, obesity and chronic inflammation.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , Inflamação/metabolismo , Neoplasias/metabolismo , Envelhecimento , Animais , Anorexia/metabolismo , Anorexia/terapia , Biomarcadores , Caquexia/metabolismo , Caquexia/terapia , Doenças Cardiovasculares/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Fator 15 de Diferenciação de Crescimento/sangue , Humanos , Camundongos , Neoplasias/imunologia
13.
Clin Cancer Res ; 15(7): 2323-34, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318483

RESUMO

PURPOSE: To test the effects of a new combination, cytosine deaminase (CD) + uracil phosphoribosyltransferase (UPRT)-mediated gene-directed enzyme prodrug therapy (GDEPT) with interleukin (IL)-12 and IL-18, on (a) growth of murine prostate and remote tumor deposits, (b) mouse survival, and (c) T helper (Th) 1/Th2 serum cytokine balance with a special focus to assess correlation with tumor burden/survival. EXPERIMENTAL DESIGN: Efficacy of intraprostatic administration of adenovirally delivered murine IL-12 and IL-18 against orthotopic RM1 tumors and lung pseudometastases was assessed in C57BL/6 mice. At necropsy, tumor growth, lung colony counts, effects on immune cell infiltration, vasculature, apoptosis, and proliferation were estimated. Next, CDUPRT-GDEPT + cytokines were tested at suboptimal doses in mice with RM1CDUPRT prostate tumors/RM1 lung deposits and analyzed as above. Effects on mouse survival were also assessed. Host immune responses to different treatments were assessed by monitoring 11 serum cytokines using Luminex technology. RESULTS: Our data show that IL-12 and IL-18, when combined with CDUPRT-GDEPT, caused significant reduction in local RM1 tumors and lung colonies with enhanced long-term survival versus individual treatments. A dramatic enhancement of tumor infiltration by a wider repertoire of immune cells and disruption of vasculature implied the combination to be more immunostimulatory and antiangiogenic. Remarkably, lowering of serum IL-4 and monocyte chemoattractant protein-1 (MCP-1) was consistently associated with lower tumor burden (local and systemic), and this, rather than an increase in Th1 cytokines, better predicted treatment efficacy. In addition, mouse survival correlated with substantially higher cytokine (Th1/Th2) levels after treatment. CONCLUSION: Locoregional application of CDUPRT-GDEPT and IL-12/IL-18 was effective against local and systemic prostate cancer and improved survival. Monitoring serum levels of IL-4 and MCP-1 may accurately reflect tumor burden and, hence, host response to therapy.


Assuntos
Citocinas/sangue , Citosina Desaminase/genética , Interleucina-12/genética , Interleucina-18/genética , Pentosiltransferases/genética , Neoplasias da Próstata/terapia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada , Flucitosina/uso terapêutico , Neoplasias Pulmonares/secundário , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pró-Fármacos/uso terapêutico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Análise de Sobrevida , Células Th2/imunologia
14.
J Immunol Methods ; 326(1-2): 41-53, 2007 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-17689554

RESUMO

Cell line-based bioassays are becoming increasingly popular for assessment of biological activities of cytokines primarily because these are easy to perform and are not subject to donor variation. A well characterised cell line with world wide availability would further minimise the inter-assay variations. C57BL/6 mice derived T cell line; CTLL-2 fits this criterion. We explored the potential of CTLL-2 cells to develop a bioassay to detection of murine (m) IL12 and mIL18. Both cytokines have shown significant activity against a number of cancers and importantly, act synergistically via mutual upregulation of each other's receptors. The preliminary flow cytometric analyses of immunostained CTLL-2 cells showed that approximately 65% expressed mIL12 and approximately 5% expressed mIL18 receptors suggesting that these may respond to mIL12. As predicted, cells incubated with different doses of mIL12 or mIL18 for 72 h were responsive to mIL12 and not to mIL18. However, when pre-treated with mIL12 for 24 h prior to incubation with mIL18, there was a significant enhancement in response. The sensitivity of the response was comparable to that obtained using the conventional splenocyte-based IFNgamma release assay. The cytokine specificity of the response was proven unequivocally when significant reduction in CTLL-2 response was observed in the presence of the relevant neutralising antibodies. Finally, we could successfully detect lowest doses of approximately 0.1 pg/microL mIL12 or 40 pg/mL of mIL18 in cell supernatants in a cytokine specific manner, which is lower than the resting levels of these cytokines in mouse sera. Again the sensitivity was comparable to that observed in the conventional IFNgamma release assay. Hence, we have demonstrated the potential of CTLL-2-based bioassay to detect biologically active mIL12 and mIL18 in biological samples accurately and reproducibly.


Assuntos
Proliferação de Células , Testes Imunológicos/métodos , Interleucina-12/análise , Interleucina-12/fisiologia , Interleucina-18/análise , Linfócitos T/imunologia , Animais , Linhagem Celular , Colorimetria , Humanos , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-12/biossíntese , Receptores de Interleucina-12/genética , Receptores de Interleucina-18/biossíntese , Receptores de Interleucina-18/genética , Linfócitos T/metabolismo , Sais de Tetrazólio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA