Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 106: 105262, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39084071

RESUMO

BACKGROUND: An estimated 1 in 350 women carry germline BRCA1/2 mutations, which confer an increased risk of developing breast and ovarian cancer, and may also contribute to subfertility. All mature, sex steroid-producing ovarian follicles are drawn from the pool of non-renewable primordial follicles, termed the 'ovarian reserve'. The clinical implications of early ovarian reserve exhaustion extend beyond infertility, to include the long-term adverse health consequences of loss of endocrine function and premature menopause. We aimed to determine whether conditional loss of Brca1 in oocytes impacts ovarian follicle numbers, oocyte quality and fertility in mice with advancing maternal age. We also aimed to determine the utility of AMH as a marker of ovarian function, by assessing circulating AMH levels in mice and women with BRCA1/2 mutations, and correlating this with ovarian follicle counts. METHODS: In this study, we addressed a longstanding question in the field regarding the functional consequences of BRCA1 inactivation in oocytes. To recapitulate loss of BRCA1 protein function in oocytes, we generated mice with conditional gene deletion of Brca1 in oocytes using Gdf9-Cre recombinase (WT: Brca1fl/flGdf9+/+; cKO: Brca1fl/flGdf9cre/+). FINDINGS: While the length of the fertile lifespan was not altered between groups after a comprehensive breeding trial, conditional loss of Brca1 in oocytes led to reduced litter size in female mice. Brca1 cKO animals had a reduced ovarian reserve and oocyte maturation was impaired with advanced maternal age at postnatal day (PN)300, compared to WT animals. Serum anti-Müllerian hormone (AMH) concentrations (the gold-standard indirect marker of the ovarian reserve used in clinical practice) were not predictive of reduced primordial follicle number in Brca1 cKO mice versus WT. Furthermore, we found no correlation between follicle number or density and serum AMH concentrations in matched samples from a small cohort of premenopausal women with BRCA1/2 mutations. INTERPRETATION: Together, our data demonstrate that BRCA1 is a key regulator of oocyte number and quality in females and suggest that caution should be used in relying on AMH as a reliable marker of the ovarian reserve in this context. FUNDING: This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIISS. This work was supported by funding from the Australian Research Council (ALW - DE21010037 and KJH - FT190100265), as well as the National Breast Cancer Foundation (IIRS-22-092) awarded to ALW and KJH. LRA, YML, LT, EOKS and MG were supported by Australian Government Research Training Program Scholarships. LRA, YML and LT were also supported by a Monash Graduate Excellence Scholarship. YC, SG and XC were supported by Monash Biomedicine Discovery Institute PhD Scholarships. LRA was also supported by a Monash University ECPF24-6809920940 Fellowship. JMS was supported by NHMRC funding (2011299). MH was supported by an NHMRC Investigator Grant (1193838).


Assuntos
Hormônio Antimülleriano , Proteína BRCA1 , Tamanho da Ninhada de Vivíparos , Oócitos , Reserva Ovariana , Animais , Oócitos/metabolismo , Feminino , Reserva Ovariana/genética , Camundongos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Hormônio Antimülleriano/sangue , Humanos , Folículo Ovariano/metabolismo , Camundongos Knockout , Técnicas de Maturação in Vitro de Oócitos
2.
Cell Genom ; 3(8): 100349, 2023 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-37601968

RESUMO

Meiotic crossovers are required for accurate chromosome segregation and producing new allelic combinations. Meiotic crossover numbers are tightly regulated within a narrow range, despite an excess of initiating DNA double-strand breaks. Here, we reveal the tumor suppressor FANCM as a meiotic anti-crossover factor in mammals. We use unique large-scale crossover analyses with both single-gamete sequencing and pedigree-based bulk-sequencing datasets to identify a genome-wide increase in crossover frequencies in Fancm-deficient mice. Gametogenesis is heavily perturbed in Fancm loss-of-function mice, which is consistent with the reproductive defects reported in humans with biallelic FANCM mutations. A portion of the gametogenesis defects can be attributed to the cGAS-STING pathway after birth. Despite the gametogenesis phenotypes in Fancm mutants, both sexes are capable of producing offspring. We propose that the anti-crossover function and role in gametogenesis of Fancm are separable and will inform diagnostic pathways for human genomic instability disorders.

3.
Nat Cancer ; 3(8): 1-13, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-36008687

RESUMO

Loss of fertility is a major concern for female reproductive-age cancer survivors, since a common side-effect of conventional cytotoxic cancer therapies is permanent damage to the ovary. While immunotherapies are increasingly becoming a standard of care for many cancers-including in the curative setting-their impacts on ovarian function and fertility are unknown. We evaluated the effect of immune checkpoint inhibitors blocking programmed cell death protein ligand 1 and cytotoxic T lymphocyte-associated antigen 4 on the ovary using tumor-bearing and tumor-free mouse models. We find that immune checkpoint inhibition increases immune cell infiltration and tumor necrosis factor-α expression within the ovary, diminishes the ovarian follicular reserve and impairs the ability of oocytes to mature and ovulate. These data demonstrate that immune checkpoint inhibitors have the potential to impair both immediate and future fertility, and studies in women should be prioritized. Additionally, fertility preservation should be strongly considered for women receiving these immunotherapies, and preventative strategies should be investigated in future studies.


Assuntos
Preservação da Fertilidade , Neoplasias , Animais , Feminino , Humanos , Inibidores de Checkpoint Imunológico , Imunoterapia/efeitos adversos , Camundongos , Oócitos/patologia
4.
J Assist Reprod Genet ; 38(6): 1405-1417, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33864208

RESUMO

PURPOSE: Genotoxic chemotherapy and radiotherapy can cause DNA double stranded breaks (DSBs) in primordial follicle (PMF) oocytes, which then undergo apoptosis. The development of effective new fertility preservation agents has been hampered, in part, by a limited understanding of DNA repair in PMF oocytes. This study investigated the induction of classical DSB repair pathways in the follicles of wild type (WT) and apoptosis-deficient Puma-/- mice in response to DSBs caused by the chemotherapy agent cisplatin. METHODS: Adult C57BL/6 WT and Puma-/- mice were injected i.p. with saline or cisplatin (5 mg/kg); ovaries were harvested at 8 or 24 h. Follicles were counted, and H2A histone family member (γH2AX) immunofluorescence used to demonstrate DSBs. DNA repair protein RAD51 homolog 1 (RAD51) and DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) immunofluorescence were used to identify DNA repair pathways utilised. RESULTS: Puma-/- mice retained 100% of follicles 24 h after cisplatin treatment. Eight hours post-treatment, γH2AX immunofluorescence showed DSBs across follicular stages in Puma-/- mice; staining returned to control levels in PMFs within 5 days, suggesting repair of PMF oocytes in this window. RAD51 immunofluorescence eight hours post-cisplatin was positive in damaged cell types in both WT and Puma-/- mice, demonstrating induction of the homologous recombination pathway. In contrast, DNA-PKcs staining were rarely observed in PMFs, indicating non-homologous end joining plays an insignificant role. CONCLUSION: PMF oocytes are able to conduct high-fidelity repair of DNA damage accumulated during chemotherapy. Therefore, apoptosis inhibition presents a viable strategy for fertility preservation in women undergoing treatment.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Cisplatino/farmacologia , Preservação da Fertilidade , Histonas/genética , Rad51 Recombinase/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/efeitos dos fármacos , Cisplatino/efeitos adversos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/genética , Proteína Quinase Ativada por DNA , Feminino , Recombinação Homóloga/efeitos dos fármacos , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Oócitos/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/patologia
5.
Toxicol Sci ; 166(1): 97-107, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30085306

RESUMO

With increasing improvements in cancer survival rates, it is critical to reduce the significant long-term side effects that afflict patients following treatment. For women, consequences of chemotherapy-induced damage to the reproductive system include infertility and premature menopause, which adversely effects cognition, mood, cardiovascular, bone, and sexual health, and increases the risk of early mortality. These long-term effects impact patient's life quality and highlight a significant and on-going burden on the health system after treatment. However, the precise mechanisms through which chemotherapeutic agents induce ovarian damage and primordial follicle depletion remain to be characterized. Hence, preventing the development of effective pharmacological methods to preserve fertility and improve quality of life after treatment. The chemotherapeutic agent 5-Fluorouracil (5FU) is not deemed cytotoxic to the ovary, however, risks to long-term fertility after multiple doses are not known. Therefore, we sought to evaluate the impact of 3, weekly doses of 5FU treatment on the ovary. Using a mouse model enabled accurate histomorphometric analysis of follicle numbers and ovarian structure and function, to accurately assess cumulative impact of 5FU on the ovary. This study clearly demonstrated that multidose 5FU treatment resulted in dramatic and progressive atresia of growing follicles and a profound decrease in ovarian volume due to reduced corpus luteum counts. However, primordial follicle numbers were unaffected. Thus, 5FU is unlikely to cause permanent infertility when administered to women of pre or reproductive age. Furthermore, this study suggests that depletion of the growing follicle population is insufficient to stimulate follicle activation and primordial follicle depletion.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Dano ao DNA , Fluoruracila/toxicidade , Atresia Folicular/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Apoptose/genética , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/crescimento & desenvolvimento , Corpo Lúteo/patologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Fluoruracila/administração & dosagem , Atresia Folicular/genética , Injeções Intraperitoneais , Camundongos Endogâmicos C57BL , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/patologia
6.
Cell Death Dis ; 9(6): 618, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29795269

RESUMO

Female gametes are stored in the ovary in structures called primordial follicles, the supply of which is non-renewable. It is well established that DNA-damaging cancer treatments can deplete the ovarian reserve of primordial follicles, causing premature ovarian failure and infertility. The precise mechanisms underlying this chemotherapy-driven follicle loss are unclear, and this has limited the development of targeted ovarian-protective agents. To address this fundamental knowledge gap, we used gene deletion mouse models to examine the role of the DNA damage-induced pro-apoptotic protein, PUMA, and its transcriptional activator TAp63, in primordial follicle depletion caused by treatment with cyclophosphamide or cisplatin. Cyclophosphamide caused almost complete destruction of the primordial follicle pool in adult wild-type (WT) mice, and a significant destructive effect was also observed for cisplatin. In striking contrast, Puma-/- mice retained 100% of their primordial follicles following either genotoxic treatment. Furthermore, elimination of PUMA alone completely preserved fertility in cyclophosphamide-treated mice, indicating that oocytes rescued from DNA damage-induced death can repair themselves sufficiently to support reproductive function and offspring health. Primordial follicles were also protected in TAp63-/- mice following cisplatin treatment, but not cyclophosphamide, suggesting mechanistic differences in the induction of apoptosis and depletion of the ovarian reserve in response to these different chemotherapies. These studies identify PUMA as a crucial effector of apoptosis responsible for depletion of primordial follicles following exposure to cyclophosphamide or cisplatin, and this indicates that inhibition of PUMA may be an effective ovarian-protective strategy during cancer treatment in women.


Assuntos
Antineoplásicos/efeitos adversos , Proteínas Reguladoras de Apoptose/deficiência , Dano ao DNA , Fertilidade , Reserva Ovariana/fisiologia , Proteínas Supressoras de Tumor/deficiência , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Cisplatino/efeitos adversos , Ciclofosfamida/efeitos adversos , Feminino , Atresia Folicular/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Reserva Ovariana/efeitos dos fármacos , Fosfoproteínas/deficiência , Fosfoproteínas/metabolismo , Transativadores/deficiência , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo
7.
Reproduction ; 151(5): 553-62, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26917450

RESUMO

Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf(-/-) mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf(-/-) and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose , Embrião de Mamíferos/patologia , Células Germinativas/patologia , Oócitos/patologia , Oogênese/fisiologia , Ovário/patologia , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Imunofluorescência , Células Germinativas/metabolismo , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/metabolismo , Ovário/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2
8.
Reproduction ; 148(2): 211-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24859845

RESUMO

The number of primordial follicles initially established within the ovary is influenced by the extent of germ cell death during foetal ovarian development, but the mechanisms that mediate this death have not been fully uncovered. In this study, we identified BBC3 (PUMA) (p53 upregulated modulator of apoptosis, also known as BCL2-binding component 3), a pro-apoptotic BH3-only protein belonging to the BCL2 family, as a critical determinant of the number of germ cells during ovarian development. Targeted disruption of the Bbc3 gene revealed a significant increase in the number of germ cells as early as embryonic day 13.5. The number of germ cells remained elevated in Bbc3(-/-) female mice compared with WT female mice throughout the remainder of embryonic and early postnatal life, resulting in a 1.9-fold increase in the number of primordial follicles in the ovary on postnatal day 10. The increase in the number of germ cells observed in the ovaries of Bbc3(-/-) mice could not be attributed to the altered proliferative activity of germ cells within the ovaries. Furthermore, BBC3 was found to be not required for the massive germ cell loss that occurs during germ cell nest breakdown. Our data indicate that BBC3 is a critical regulator of germ cell death that acts during the migratory phase of oogenesis or very soon after the arrival of germ cells in the gonad and that BBC3-mediated cell death limits the number of primordial follicles established in the initial ovarian reserve.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose , Embrião de Mamíferos/citologia , Células Germinativas/patologia , Folículo Ovariano/patologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Células Germinativas/metabolismo , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Folículo Ovariano/metabolismo
9.
Cancer Res ; 70(7): 2665-74, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20332236

RESUMO

Several reports have shown that prophylactic depletion of regulatory T cells (Treg) using various monoclonal antibodies (mAb) in mice can stimulate potent antitumor immune responses and prevent tumor development. These same depletion methods do not significantly suppress tumor growth in a therapeutic setting. Although different strategies to deplete FoxP3(+) Treg have been used, no study has systematically compared these qualitatively for the effector mechanisms they each liberate. Herein, using prophylactic depletion of FoxP3(+) Tregs with either anti-CD4, anti-CD25, or anti-FR4 mAbs, we have compared the cellular and effector requirements for elimination of the renal carcinoma RENCA and prevention of methylcholanthrene-induced fibrosarcoma. Collectively from these two models, it was clear that CD8(+) T cells and natural killer cells played an important role downstream of Treg depletion. However, whereas all three mAbs quantitatively depleted FoxP3(+) T cells to a similar extent, subtle differences in the downstream mechanisms of tumor control existed for all three approaches. In general, neutralization of any lymphocyte subset or effector mechanism was insufficient to alter tumor suppression initiated by Treg depletion, and in some settings, the neutralization of multiple effector mechanisms failed to prevent tumor rejection. These studies reveal that Tregs control multiple redundant elements of the immune effector response capable of inhibiting tumor initiation and underscore the importance of effectively targeting these cells in any cancer immunotherapy.


Assuntos
Adenocarcinoma/imunologia , Fibrossarcoma/imunologia , Neoplasias Renais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos CD4/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citocinas/imunologia , Fatores de Transcrição Forkhead/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Metilcolantreno , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/imunologia
10.
Nature ; 450(7171): 903-7, 2007 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-18026089

RESUMO

The capacity of immunity to control and shape cancer, that is, cancer immunoediting, is the result of three processes that function either independently or in sequence: elimination (cancer immunosurveillance, in which immunity functions as an extrinsic tumour suppressor in naive hosts); equilibrium (expansion of transformed cells is held in check by immunity); and escape (tumour cell variants with dampened immunogenicity or the capacity to attenuate immune responses grow into clinically apparent cancers). Extensive experimental support now exists for the elimination and escape processes because immunodeficient mice develop more carcinogen-induced and spontaneous cancers than wild-type mice, and tumour cells from immunodeficient mice are more immunogenic than those from immunocompetent mice. In contrast, the equilibrium process was inferred largely from clinical observations, including reports of transplantation of undetected (occult) cancer from organ donor into immunosuppressed recipients. Herein we use a mouse model of primary chemical carcinogenesis and demonstrate that equilibrium occurs, is mechanistically distinguishable from elimination and escape, and that neoplastic cells in equilibrium are transformed but proliferate poorly in vivo. We also show that tumour cells in equilibrium are unedited but become edited when they spontaneously escape immune control and grow into clinically apparent tumours. These results reveal that, in addition to destroying tumour cells and sculpting tumour immunogenicity, the immune system of a naive mouse can also restrain cancer growth for extended time periods.


Assuntos
Neoplasia Residual/imunologia , Sarcoma/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Progressão da Doença , Feminino , Genes RAG-1/genética , Imunidade Ativa/efeitos dos fármacos , Imunidade Ativa/imunologia , Imunocompetência/imunologia , Interferon gama/imunologia , Masculino , Metilcolantreno , Camundongos , Modelos Imunológicos , Neoplasia Residual/induzido quimicamente , Neoplasia Residual/patologia , Sarcoma/induzido quimicamente , Sarcoma/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
11.
Cancer Res ; 67(11): 5454-60, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545627

RESUMO

The concept of tumor immune surveillance has been supported by several recent studies in mice which show that immune effector mechanisms suppress hematologic malignancy. However, because the most common forms of human cancer are epithelial in origin, and comparatively very little data supports the immune surveillance of epithelial malignancies, we have chosen to evaluate the role of perforin-mediated cytotoxicity in the prevention of BALB/c Her2/neu-induced mammary cancer. Interestingly, perforin significantly delayed the onset of mammary tumorigenesis and reduced the number of mammary tumors without improving survival. Natural killer cell, but not CD8+ T cell, depletion resulted in a similar phenotype to perforin deficiency in this regard. Histologic analysis further indicated that the effect of perforin was most evident during the earliest stages of carcinogenesis rather than prior to or during the hyperplastic phase. This data suggests that perforin may mediate some suppression of epithelial carcinogenesis by intervening early in the tumor development process.


Assuntos
Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Glicoproteínas de Membrana/imunologia , Oncogenes , Proteínas Citotóxicas Formadoras de Poros/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Feminino , Células Matadoras Naturais/imunologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Perforina , Proteínas Citotóxicas Formadoras de Poros/deficiência , Proteínas Citotóxicas Formadoras de Poros/genética , Receptor ErbB-2/deficiência , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia
12.
J Immunol ; 178(12): 7540-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548588

RESUMO

This study demonstrates that type I IFNs are an early and critical regulator of NK cell numbers, activation, and antitumor activity. Using both IFNAR1- and IFNAR2-deficient mice, as well as an IFNAR1-blocking Ab, we demonstrate that endogenous type I IFN is critical for controlling NK cell-mediated antitumor responses in many experimental tumor models, including protection from methylcholanthrene-induced sarcomas, resistance to the NK cell-sensitive RMA-S tumor and cytokine immunotherapy of lung metastases. Protection from RMA-S afforded by endogenous type I IFN is more potent than that of other effector molecules such as IFN-gamma, IL-12, IL-18, and perforin. Furthermore, cytokine immunotherapy using IL-12, IL-18, or IL-21 was effective in the absence of endogenous type I IFN, however the antimetastatic activity of IL-2 was abrogated in IFNAR-deficient mice, primarily due to a defect in IL-2-induced cytotoxic activity. This study demonstrates that endogenous type I IFN is a central mediator of NK cell antitumor responses.


Assuntos
Interferon Tipo I/fisiologia , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Animais , Anticorpos/farmacologia , Citocinas/uso terapêutico , Homeostase , Imunoterapia , Interferon Tipo I/uso terapêutico , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Camundongos , Camundongos Mutantes , Neoplasias/prevenção & controle , Neoplasias/terapia , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Immunol ; 176(10): 6347-55, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16670347

RESUMO

Tumor cell apoptosis is the basis of many cancer therapies, and tumor-specific T cells are the principal effectors of successful anti-tumor immunotherapies. In this study, we show that induction of tumor cell apoptosis by agonistic mAb against DR5, combined with delayed IL-21 treatment, suppressed tumor growth and pre-established tumor metastases. Synergistic effects of the combination were observed in several tumor models where the target tumor was sensitive to DR5-mediated apoptosis. IL-21 promoted tumor-specific CTL activity and enhanced memory responses to tumor rechallenge. These results indicate that a rational combination of Ab-based therapy that causes tumor cell apoptosis and a cytokine that promotes T cell memory is a useful new strategy for cancer immunotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citotoxicidade Imunológica , Ativação Linfocitária/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Linhagem Celular Tumoral , Interleucinas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Linfócitos T Citotóxicos/metabolismo
14.
J Immunol ; 175(9): 5586-90, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16237043

RESUMO

TNF apoptosis-inducing ligand is attracting considerable interest as a potential extrinsic tumor suppressor mechanism, although previous reports have conveyed somewhat contrasting views regarding the likely importance of this pathway. In this study, we provide the first evaluation of spontaneous tumor formation over the life span of TRAIL-deficient mice. Interestingly, >25% of these mice do develop lymphoid malignancies after 500 days of life. TRAIL suppressed the initiation and development of both tumors of lymphoid and stromal origin in the context of the loss of at least one p53 allele. Specific examination of the role of TRAIL in Her2/neu oncogene-driven mammary epithelial cancer revealed no critical role for TRAIL despite the inherent TRAIL sensitivity of such mammary carcinomas. Overall, the data indicate an important function of TRAIL in controlling carcinogenesis, but suggest that further examination of this pathway in epithelial malignancies is warranted.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Neoplasias Hematológicas/etiologia , Glicoproteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Genes erbB-2 , Genes p53 , Linfoma/etiologia , Linfoma/prevenção & controle , Neoplasias Mamárias Animais/etiologia , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/deficiência
15.
J Exp Med ; 202(5): 583-8, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16129707

RESUMO

The activation NKG2D receptor has been shown to play an important role in the control of experimental tumor growth and metastases expressing ligands for NKG2D; however, a function for this recognition pathway in host protection from de novo tumorigenesis has never been demonstrated. We show that neutralization of NKG2D enhances the sensitivity of wild-type (WT) C57BL/6 and BALB/c mice to methylcholanthrene (MCA)-induced fibrosarcoma. The importance of the NKG2D pathway was additionally illustrated in mice deficient for either IFN-gamma or tumor necrosis factor-related apoptosis-inducing ligand, whereas mice depleted of natural killer cells, T cells, or deficient for perforin did not display any detectable NKG2D phenotype. Furthermore, IL-12 therapy preventing MCA-induced sarcoma formation was also largely dependent on the NKG2D pathway. Although NKG2D ligand expression was variable or absent on sarcomas emerging in WT mice, sarcomas derived from perforin-deficient mice were Rae-1(+) and immunogenic when transferred into WT syngeneic mice. These findings suggest an important early role for the NKG2D in controlling and shaping tumor formation.


Assuntos
Fibrossarcoma/imunologia , Fibrossarcoma/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/imunologia , Animais , Anticorpos Monoclonais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas de Transporte/metabolismo , Fibrossarcoma/induzido quimicamente , Fibrossarcoma/tratamento farmacológico , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/metabolismo , Interleucina-12/metabolismo , Interleucina-12/uso terapêutico , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Glicoproteínas de Membrana/deficiência , Proteínas de Membrana , Metilcolantreno , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Testes de Neutralização , Perforina , Proteínas Citotóxicas Formadoras de Poros , Receptores de Células Matadoras Naturais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA