Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
EBioMedicine ; 106: 105262, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39084071

RESUMEN

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).


Asunto(s)
Hormona Antimülleriana , Proteína BRCA1 , Tamaño de la Camada , Oocitos , Reserva Ovárica , Animales , Oocitos/metabolismo , Femenino , Reserva Ovárica/genética , Ratones , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Hormona Antimülleriana/sangre , Humanos , Folículo Ovárico/metabolismo , Ratones Noqueados , Técnicas de Maduración In Vitro de los Oocitos
2.
Front Endocrinol (Lausanne) ; 14: 1268009, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37900135

RESUMEN

Introduction: The non-growing, meiotically-arrested oocytes housed within primordial follicles are exquisitely sensitive to genotoxic insults from endogenous and exogenous sources. Even a single DNA double-strand break (DSB) can trigger oocyte apoptosis, which can lead to accelerated depletion of the ovarian reserve, early loss of fertility and menopause. Therefore, repair of DNA damage is important for preserving the quality of oocytes to sustain fertility across the reproductive lifespan. This study aimed to evaluate the role of KU80 (encoded by the XRCC5 gene) - an essential component of the non-homologous end joining (NHEJ) pathway - in the repair of oocyte DNA DSBs during reproductive ageing, and following insult caused by the DNA-damaging chemotherapies cyclophosphamide and cisplatin. Methods: To investigate the importance of KU80 following endogenous and exogenous DNA damage, ovaries from conditional oocyte-specific Xrcc5 knockout (Xrcc5 cKO) and wildtype (WT) mice that were aged or exposed to DNA damage-inducing chemotherapy were compared. Ovarian follicles and oocytes were quantified, morphologically assessed and analysed via immunohistochemistry for markers of DNA damage and apoptosis. In addition, chemotherapy exposed mice were superovulated, and the numbers and quality of mature metaphase- II (MII) oocytes were assessed. Results: The number of healthy follicles, atretic (dying) follicles, and corpora lutea were similar in Xrcc5 cKO and WT mice at PN50, PN200 and PN300. Additionally, primordial follicle number and ovulation rates were similar in young adult Xrcc5 cKO and WT mice following treatment with cyclophosphamide (75mg/kg), cisplatin (4mg/kg), or vehicle control (saline). Furthermore, KU80 was not essential for the repair of exogenously induced DNA damage in primordial follicle oocytes. Discussion: These data indicate that KU80 is not required for maintenance of the ovarian reserve, follicle development, or ovulation during maternal ageing. Similarly, this study also indicates that KU80 is not required for the repair of exogenously induced DSBs in the prophase-arrested oocytes of primordial follicles.


Asunto(s)
Cisplatino , Autoantígeno Ku , Folículo Ovárico , Animales , Femenino , Ratones , Ciclofosfamida/farmacología , ADN , Oocitos/fisiología , Folículo Ovárico/fisiología , Profase , Autoantígeno Ku/genética
3.
Cell Genom ; 3(8): 100349, 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37601968

RESUMEN

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.

4.
Cell Death Dis ; 14(7): 397, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407587

RESUMEN

Mammalian oocytes spend most of their life in a unique state of cell cycle arrest at meiotic prophase I, during which time they are exposed to countless DNA-damaging events. Recent studies have shown that DNA double-strand break repair occurs predominantly via the homologous recombination (HR) pathway in small non-growing meiotically arrested oocytes (primordial follicle stage). However, the DNA repair mechanisms employed by fully grown meiotically arrested oocytes (GV-stage) have not been studied in detail. Here we established a conditional knockout mouse model to explore the role of Ku80, a critical component of the nonhomologous end joining (NHEJ) pathway, in the repair of DNA damage in GV oocytes. GV oocytes lacking Ku80 failed to repair etoposide-induced DNA damage, even when only low levels of damage were sustained. This indicates Ku80 is needed to resolve DSBs and that HR cannot compensate for a compromised NHEJ pathway in fully-grown oocytes. When higher levels of DNA damage were induced, a severe delay in M-phase entry was observed in oocytes lacking XRCC5 compared to wild-type oocytes, suggesting that Ku80-dependent repair of DNA damage is important for the timely release of oocytes from prophase I and resumption of meiosis. Ku80 was also found to be critical for chromosome integrity during meiotic maturation following etoposide exposure. These data demonstrate that Ku80, and NHEJ, are vital for quality control in mammalian GV stage oocytes and reveal that DNA repair pathway choice differs in meiotically arrested oocytes according to growth status.


Asunto(s)
Meiosis , Oocitos , Animales , Ratones , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Etopósido/farmacología , Mamíferos , Oocitos/metabolismo
5.
Cells ; 12(4)2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36831314

RESUMEN

A common herbicide, atrazine, is associated with poor health. Atrazine acts as an endocrine disruptor at supra-environmental levels. Little research, however, has been conducted regarding chronic exposure to environmental atrazine concentrations across generations. This study utilized comprehensive endpoint measures to investigate the effects of chronic exposure to a conservative atrazine concentration (0.02 ng/mL), measured in Australian waterways, on male mice fertility across two generations. Mice were exposed through the maternal line, from the pre-conception period and through the F1 and F2 generations until three or six months of age. Atrazine did not impact sperm function, testicular morphology nor germ cell parameters but did alter the expression of steroidogenic genes in the F1, down-regulating the expression of Cyp17a1 (Cytochrome P450 family 17, subfamily A member 1; p = 0.0008) and Ddx4 (DEAD-box helicase 4; p = 0.007), and up-regulating the expression of Star (Steroidogenic acute regulatory protein; p = 0.017). In the F2, atrazine induced up-regulation in the expression of Star (p = 0.016). The current study demonstrates that chronic exposure to an environmentally relevant atrazine concentration perturbs testicular steroid-associated gene expression that varies across generations. Future studies through the paternal and combined parental lineages should be undertaken to further elucidate the multigenerational effects of atrazine on male fertility.


Asunto(s)
Atrazina , Herbicidas , Masculino , Ratones , Animales , Atrazina/farmacología , Australia , Semen , Herbicidas/farmacología , Testículo
6.
Nat Cancer ; 3(8): 1-13, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36008687

RESUMEN

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.


Asunto(s)
Preservación de la Fertilidad , Neoplasias , Animales , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia/efectos adversos , Ratones , Oocitos/patología
7.
J Assist Reprod Genet ; 38(6): 1405-1417, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33864208

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Cisplatino/farmacología , Preservación de la Fertilidad , Histonas/genética , Recombinasa Rad51/genética , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Cisplatino/efectos adversos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/genética , Proteína Quinasa Activada por ADN , Femenino , Recombinación Homóloga/efectos de los fármacos , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/prevención & control , Oocitos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/patología
8.
Proc Natl Acad Sci U S A ; 117(21): 11513-11522, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32381741

RESUMEN

Female fertility and offspring health are critically dependent on an adequate supply of high-quality oocytes, the majority of which are maintained in the ovaries in a unique state of meiotic prophase arrest. While mechanisms of DNA repair during meiotic recombination are well characterized, the same is not true for prophase-arrested oocytes. Here we show that prophase-arrested oocytes rapidly respond to γ-irradiation-induced DNA double-strand breaks by activating Ataxia Telangiectasia Mutated, phosphorylating histone H2AX, and localizing RAD51 to the sites of DNA damage. Despite mobilizing the DNA repair response, even very low levels of DNA damage result in the apoptosis of prophase-arrested oocytes. However, we show that, when apoptosis is inhibited, severe DNA damage is corrected via homologous recombination repair. The repair is sufficient to support fertility and maintain health and genetic fidelity in offspring. Thus, despite the preferential induction of apoptosis following exogenously induced genotoxic stress, prophase-arrested oocytes are highly capable of functionally efficient DNA repair. These data implicate DNA repair as a key quality control mechanism in the female germ line and a critical determinant of fertility and genetic integrity.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Fertilidad/fisiología , Oocitos/fisiología , Animales , Apoptosis/fisiología , Femenino , Masculino , Ratones Endogámicos C57BL , Profase/fisiología
9.
Toxicol Sci ; 166(1): 97-107, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085306

RESUMEN

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.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Daño del ADN , Fluorouracilo/toxicidad , Atresia Folicular/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Apoptosis/genética , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/crecimiento & desarrollo , Cuerpo Lúteo/patología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Fluorouracilo/administración & dosificación , Atresia Folicular/genética , Inyecciones Intraperitoneales , Ratones Endogámicos C57BL , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/patología
10.
Cell Death Dis ; 9(6): 618, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29795269

RESUMEN

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.


Asunto(s)
Antineoplásicos/efectos adversos , Proteínas Reguladoras de la Apoptosis/deficiencia , Daño del ADN , Fertilidad , Reserva Ovárica/fisiología , Proteínas Supresoras de Tumor/deficiencia , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Cisplatino/efectos adversos , Ciclofosfamida/efectos adversos , Femenino , Atresia Folicular/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/patología , Reserva Ovárica/efectos de los fármacos , Fosfoproteínas/deficiencia , Fosfoproteínas/metabolismo , Transactivadores/deficiencia , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo
11.
Reproduction ; 151(5): 553-62, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26917450

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Apoptosis , Embrión de Mamíferos/patología , Células Germinativas/patología , Oocitos/patología , Oogénesis/fisiología , Ovario/patología , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Ovario/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2
12.
Reproduction ; 148(2): 211-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24859845

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis , Embrión de Mamíferos/citología , Células Germinativas/patología , Folículo Ovárico/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Folículo Ovárico/metabolismo
13.
Cancer Res ; 70(7): 2665-74, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20332236

RESUMEN

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.


Asunto(s)
Adenocarcinoma/inmunología , Fibrosarcoma/inmunología , Neoplasias Renales/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD4/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citocinas/inmunología , Factores de Transcripción Forkhead/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Metilcolantreno , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Superficie Celular/inmunología
14.
Nature ; 450(7171): 903-7, 2007 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-18026089

RESUMEN

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.


Asunto(s)
Neoplasia Residual/inmunología , Sarcoma/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Genes RAG-1/genética , Inmunidad Activa/efectos de los fármacos , Inmunidad Activa/inmunología , Inmunocompetencia/inmunología , Interferón gamma/inmunología , Masculino , Metilcolantreno , Ratones , Modelos Inmunológicos , Neoplasia Residual/inducido químicamente , Neoplasia Residual/patología , Sarcoma/inducido químicamente , Sarcoma/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
15.
Cancer Res ; 67(11): 5454-60, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17545627

RESUMEN

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.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Glicoproteínas de Membrana/inmunología , Oncogenes , Proteínas Citotóxicas Formadoras de Poros/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Femenino , Células Asesinas Naturales/inmunología , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Perforina , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Proteínas Citotóxicas Formadoras de Poros/genética , Receptor ErbB-2/deficiencia , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología
16.
J Immunol ; 178(12): 7540-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548588

RESUMEN

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.


Asunto(s)
Interferón Tipo I/fisiología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Anticuerpos/farmacología , Citocinas/uso terapéutico , Homeostasis , Inmunoterapia , Interferón Tipo I/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos , Ratones , Ratones Mutantes , Neoplasias/prevención & control , Neoplasias/terapia , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Immunol ; 176(10): 6347-55, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16670347

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citotoxicidad Inmunológica , Activación de Linfocitos/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T Citotóxicos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Línea Celular Tumoral , Interleucinas/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Linfocitos T Citotóxicos/metabolismo
18.
J Immunol ; 175(9): 5586-90, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16237043

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Neoplasias Hematológicas/etiología , Glicoproteínas de Membrana/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Genes erbB-2 , Genes p53 , Linfoma/etiología , Linfoma/prevención & control , Neoplasias Mamarias Animales/etiología , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/deficiencia
19.
J Exp Med ; 202(5): 583-8, 2005 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16129707

RESUMEN

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.


Asunto(s)
Fibrosarcoma/inmunología , Fibrosarcoma/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/inmunología , Animales , Anticuerpos Monoclonales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Portadoras/metabolismo , Fibrosarcoma/inducido químicamente , Fibrosarcoma/tratamiento farmacológico , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/metabolismo , Interleucina-12/metabolismo , Interleucina-12/uso terapéutico , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Glicoproteínas de Membrana/deficiencia , Proteínas de la Membrana , Metilcolantreno , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK , Pruebas de Neutralización , Perforina , Proteínas Citotóxicas Formadoras de Poros , Receptores de Células Asesinas Naturales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA