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








Base de dados
Intervalo de ano de publicação
2.
Nature ; 589(7842): 442-447, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33361811

RESUMO

Successful pregnancies rely on adaptations within the mother1, including marked changes within the immune system2. It has long been known that the thymus, the central lymphoid organ, changes markedly during pregnancy3. However, the molecular basis and importance of this process remain largely obscure. Here we show that the osteoclast differentiation receptor RANK4,5 couples female sex hormones to the rewiring of the thymus during pregnancy. Genetic deletion of Rank (also known as Tnfrsf11a) in thymic epithelial cells results in impaired thymic involution and blunted expansion of natural regulatory T (Treg) cells in pregnant female mice. Sex hormones, in particular progesterone, drive the development of thymic Treg cells through RANK in a manner that depends on AIRE+ medullary thymic epithelial cells. The depletion of Rank in the mouse thymic epithelium results in reduced accumulation of natural Treg cells in the placenta, and an increase in the number of miscarriages. Thymic deletion of Rank also results in impaired accumulation of Treg cells in visceral adipose tissue, and is associated with enlarged adipocyte size, tissue inflammation, enhanced maternal glucose intolerance, fetal macrosomia, and a long-lasting transgenerational alteration in glucose homeostasis, which are all key hallmarks of gestational diabetes. Transplantation of Treg cells rescued fetal loss, maternal glucose intolerance and fetal macrosomia. In human pregnancies, we found that gestational diabetes also correlates with a reduced number of Treg cells in the placenta. Our findings show that RANK promotes the hormone-mediated development of thymic Treg cells during pregnancy, and expand the functional role of maternal Treg cells to the development of gestational diabetes and the transgenerational metabolic rewiring of glucose homeostasis.


Assuntos
Diabetes Gestacional/imunologia , Morte Fetal/etiologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Linfócitos T Reguladores/imunologia , Timo/imunologia , Adipócitos/patologia , Animais , Proliferação de Células , Diabetes Gestacional/etiologia , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patologia , Células Epiteliais/imunologia , Feminino , Feto/imunologia , Feto/metabolismo , Feto/patologia , Glucose/metabolismo , Intolerância à Glucose/genética , Humanos , Gordura Intra-Abdominal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Placenta/imunologia , Placenta/patologia , Gravidez , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Linfócitos T Reguladores/citologia , Timo/citologia , Fatores de Transcrição/metabolismo , Proteína AIRE
3.
Int J Gynecol Cancer ; 30(11): 1667-1671, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33033166

RESUMO

BACKGROUND: On March 16, 2020, the federal government of Austria declared a nationwide lockdown due to the COVID-19 pandemic. Since the lockdown, screening examinations and routine checkups have been restricted to prevent the spread of the virus and to increase the hospitals' bed capacity across the country. This resulted in a severe decline of patient referrals to the hospitals. OBJECTIVE: To assess the impact of the COVID-19 pandemic on the rate of newly diagnosed gynecological and breast cancers in Austria. METHODS: Data of 2077 patients from 18 centers in Austria with newly diagnosed gynecological or breast cancer between January and May 2019 and January and May 2020 were collected. Clinical parameters, including symptoms, performance status, co-morbidities, and referral status, were compared between the time before and after the COVID-19 outbreak. RESULTS: Our results showed a slight increase of newly diagnosed cancers in January and February 2020 as compared with 2019 (+2 and +35%, respectively) and a strong decline in newly diagnosed tumors since the lockdown: -24% in March 2020 versus March 2019, -49% in April 2020 versus April 2019, -49% in May 2020 versus May 2019. Two-thirds of patients diagnosed during the pandemic presented with tumor-specific symptoms compared with less than 50% before the pandemic (p<0.001). Moreover, almost 50% of patients in 2020 had no co-morbidities compared with 35% in 2019 (p<0.001). Patients, who already had a malignant disease, were rarely diagnosed with a new cancer in 2020 as compared with 2019 (11% vs 6%; p<0.001). CONCLUSIONS: The lockdown led to a decreased number of newly diagnosed gynecological and breast cancers. The decreased accessibility of the medical services and postponed diagnosis of potentially curable cancers during the COVID-19 pandemic may be a step backwards in our healthcare system and might impair cancer treatment outcomes. Therefore, new strategies to manage early cancer detection are needed to optimize cancer care in a time of pandemic in the future.


Assuntos
Neoplasias da Mama/epidemiologia , Infecções por Coronavirus , Neoplasias dos Genitais Femininos/epidemiologia , Pandemias , Pneumonia Viral , Adulto , Idoso , Idoso de 80 Anos ou mais , Áustria/epidemiologia , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/terapia , COVID-19 , Feminino , Neoplasias dos Genitais Femininos/diagnóstico , Neoplasias dos Genitais Femininos/terapia , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
4.
Trends Cell Biol ; 28(3): 213-223, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29241686

RESUMO

The tumor necrosis factor (TNF) receptor RANK (TNFRSF11A) and its ligand RANKL (TNFSF11) regulate osteoclast development and bone metabolism. They also control stem cell expansion and proliferation of mammary epithelial cells via the sex hormone progesterone. As such, RANKL and RANK have been implicated in the onset of hormone-induced breast cancer. Recently, RANK/RANKL were identified as crucial regulators for BRCA1 mutation-driven breast cancer. Current prevention strategies for BRCA1 mutation carriers are associated with wide-ranging risks; therefore, the search for alternative, non-invasive strategies is of paramount importance. We summarize here the functions of the RANKL/RANK pathway in mammalian physiology and focus on its recently uncovered role in breast cancer. We propose that anti-RANKL therapy should be pursued as a preventative strategy for breast cancer.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Feminino , Humanos , Mutação , Células-Tronco Neoplásicas/metabolismo , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores
5.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29118048

RESUMO

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Células Epiteliais Alveolares/metabolismo , Animais , Respiração Celular , Células Cultivadas , Metabolismo Energético , Feminino , Hormônios Esteroides Gonadais/fisiologia , Homeostase , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Mucosa Respiratória/metabolismo
6.
Elife ; 62017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28806168

RESUMO

To determine the dynamics of allelic-specific expression during mouse development, we analyzed RNA-seq data from 23 F1 tissues from different developmental stages, including 19 female tissues allowing X chromosome inactivation (XCI) escapers to also be detected. We demonstrate that allelic expression arising from genetic or epigenetic differences is highly tissue-specific. We find that tissue-specific strain-biased gene expression may be regulated by tissue-specific enhancers or by post-transcriptional differences in stability between the alleles. We also find that escape from X-inactivation is tissue-specific, with leg muscle showing an unexpectedly high rate of XCI escapers. By surveying a range of tissues during development, and performing extensive validation, we are able to provide a high confidence list of mouse imprinted genes including 18 novel genes. This shows that cluster size varies dynamically during development and can be substantially larger than previously thought, with the Igf2r cluster extending over 10 Mb in placenta.


Assuntos
Alelos , Regulação da Expressão Gênica no Desenvolvimento , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Camundongos , Análise de Sequência de RNA , Inativação do Cromossomo X
7.
Open Biol ; 6(11)2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27881737

RESUMO

RANK and RANKL, a receptor ligand pair belonging to the tumour necrosis factor family, are the critical regulators of osteoclast development and bone metabolism. Besides their essential function in bone, RANK and RANKL have also been identified as the key factors for the formation of a lactating mammary gland in pregnancy. Mechanistically, RANK and RANKL link the sex hormone progesterone with stem cell expansion and proliferation of mammary epithelial cells. Based on their normal physiology, RANKL/RANK control the onset of hormone-induced breast cancer through the expansion of mammary progenitor cells. Recently, we and others were able to show that RANK and RANKL are also critical regulators of BRCA1-mutation-driven breast cancer. Currently, the preventive strategy for BRCA1-mutation carriers includes preventive mastectomy, associated with wide-ranging risks and psychosocial effects. The search for an alternative non-invasive prevention strategy is therefore of paramount importance. As our work strongly implicates RANK and RANKL as key molecules involved in the initiation of BRCA1-associated breast cancer, we propose that anti-RANKL therapy could be a feasible preventive strategy for women carrying BRCA1 mutations, and by extension to other women with high risk of breast cancer.


Assuntos
Neoplasias da Mama/prevenção & controle , Osteoporose/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Proteína BRCA1/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ensaios Clínicos como Assunto , Denosumab/farmacologia , Denosumab/uso terapêutico , Feminino , Humanos , Mastectomia , Mutação , Ligante RANK/antagonistas & inibidores
8.
Cancer Cell ; 30(1): 147-160, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27411589

RESUMO

Caloric restriction mimetics (CRMs) mimic the biochemical effects of nutrient deprivation by reducing lysine acetylation of cellular proteins, thus triggering autophagy. Treatment with the CRM hydroxycitrate, an inhibitor of ATP citrate lyase, induced the depletion of regulatory T cells (which dampen anticancer immunity) from autophagy-competent, but not autophagy-deficient, mutant KRAS-induced lung cancers in mice, thereby improving anticancer immunosurveillance and reducing tumor mass. Short-term fasting or treatment with several chemically unrelated autophagy-inducing CRMs, including hydroxycitrate and spermidine, improved the inhibition of tumor growth by chemotherapy in vivo. This effect was only observed for autophagy-competent tumors, depended on the presence of T lymphocytes, and was accompanied by the depletion of regulatory T cells from the tumor bed.


Assuntos
Citratos/administração & dosagem , Neoplasias Experimentais/dietoterapia , Neoplasias Experimentais/tratamento farmacológico , Espermidina/administração & dosagem , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Autofagia , Proteína 5 Relacionada à Autofagia/genética , Restrição Calórica/métodos , Linhagem Celular Tumoral , Citratos/farmacologia , Humanos , Metotrexato/administração & dosagem , Metotrexato/farmacologia , Camundongos , Monitorização Imunológica , Mutação , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Espermidina/farmacologia
9.
Cell Res ; 26(7): 761-74, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27241552

RESUMO

Breast cancer is the most common female cancer, affecting approximately one in eight women during their life-time. Besides environmental triggers and hormones, inherited mutations in the breast cancer 1 (BRCA1) or BRCA2 genes markedly increase the risk for the development of breast cancer. Here, using two different mouse models, we show that genetic inactivation of the key osteoclast differentiation factor RANK in the mammary epithelium markedly delayed onset, reduced incidence, and attenuated progression of Brca1;p53 mutation-driven mammary cancer. Long-term pharmacological inhibition of the RANK ligand RANKL in mice abolished the occurrence of Brca1 mutation-driven pre-neoplastic lesions. Mechanistically, genetic inactivation of Rank or RANKL/RANK blockade impaired proliferation and expansion of both murine Brca1;p53 mutant mammary stem cells and mammary progenitors from human BRCA1 mutation carriers. In addition, genome variations within the RANK locus were significantly associated with risk of developing breast cancer in women with BRCA1 mutations. Thus, RANKL/RANK control progenitor cell expansion and tumorigenesis in inherited breast cancer. These results present a viable strategy for the possible prevention of breast cancer in BRCA1 mutant patients.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Proteína BRCA2/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligante RANK/antagonistas & inibidores , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptores de Progesterona/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Cytokine Growth Factor Rev ; 25(2): 205-14, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24486161

RESUMO

RANK and its ligand RANKL are key molecules in bone metabolism and are critically involved in pathologic bone disorders. Deregulation of the RANK/RANKL system is for example a main reason for the development of postmenopausal osteoporosis, which affects millions of women worldwide. Another essential function of RANK and RANKL is the development of a functional lactating mammary gland during pregnancy. Sex hormones, in particular progesterone, induce RANKL expression resulting in proliferation of mammary epithelial cells. Moreover, RANK and RANKL have been shown to regulate mammary epithelial stem cells. RANK and RANKL were also identified as critical mechanism in the development of hormone-induced breast cancer and metastatic spread to bone. In this review, we will focus on the various RANK/RANKL functions ranging from bone physiology, immune regulation, and initiation of breast cancer.


Assuntos
Osso e Ossos/fisiologia , Osteoprotegerina/fisiologia , Ligante RANK/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Animais , Doenças Autoimunes/genética , Doenças Ósseas/metabolismo , Neoplasias Ósseas/patologia , Osso e Ossos/metabolismo , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Feminino , Humanos , Camundongos , NF-kappa B/genética , Osteoporose , Osteoprotegerina/genética
11.
Nat Commun ; 5: 3056, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24445999

RESUMO

Autophagy is a mechanism by which starving cells can control their energy requirements and metabolic states, thus facilitating the survival of cells in stressful environments, in particular in the pathogenesis of cancer. Here we report that tissue-specific inactivation of Atg5, essential for the formation of autophagosomes, markedly impairs the progression of KRas(G12D)-driven lung cancer, resulting in a significant survival advantage of tumour-bearing mice. Autophagy-defective lung cancers exhibit impaired mitochondrial energy homoeostasis, oxidative stress and a constitutively active DNA damage response. Genetic deletion of the tumour suppressor p53 reinstates cancer progression of autophagy-deficient tumours. Although there is improved survival, the onset of Atg5-mutant KRas(G12D)-driven lung tumours is markedly accelerated. Mechanistically, increased oncogenesis maps to regulatory T cells. These results demonstrate that, in KRas(G12D)-driven lung cancer, Atg5-regulated autophagy accelerates tumour progression; however, autophagy also represses early oncogenesis, suggesting a link between deregulated autophagy and regulatory T cell controlled anticancer immunity.


Assuntos
Autofagia/fisiologia , Modelos Animais de Doenças , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/fisiopatologia , Proteínas Associadas aos Microtúbulos/fisiologia , Animais , Proteína 5 Relacionada à Autofagia , Progressão da Doença , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia
12.
Science ; 337(6102): 1678-84, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-23019653

RESUMO

Cancer cells accommodate multiple genetic and epigenetic alterations that initially activate intrinsic (cell-autonomous) and extrinsic (immune-mediated) oncosuppressive mechanisms. Only once these barriers to oncogenesis have been overcome can malignant growth proceed unrestrained. Tetraploidization can contribute to oncogenesis because hyperploid cells are genomically unstable. We report that hyperploid cancer cells become immunogenic because of a constitutive endoplasmic reticulum stress response resulting in the aberrant cell surface exposure of calreticulin. Hyperploid, calreticulin-exposing cancer cells readily proliferated in immunodeficient mice and conserved their increased DNA content. In contrast, hyperploid cells injected into immunocompetent mice generated tumors only after a delay, and such tumors exhibited reduced DNA content, endoplasmic reticulum stress, and calreticulin exposure. Our results unveil an immunosurveillance system that imposes immunoselection against hyperploidy in carcinogen- and oncogene-induced cancers.


Assuntos
Estresse do Retículo Endoplasmático/imunologia , Vigilância Imunológica , Neoplasias/genética , Neoplasias/imunologia , Ploidias , Animais , Calreticulina/imunologia , Linhagem Celular Tumoral , Imunodeficiência de Variável Comum/genética , DNA de Neoplasias/análise , DNA de Neoplasias/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Imunocompetência , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/induzido quimicamente , Fosforilação
13.
Nature ; 487(7408): 477-81, 2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-22837003

RESUMO

Malnutrition affects up to one billion people in the world and is a major cause of mortality. In many cases, malnutrition is associated with diarrhoea and intestinal inflammation, further contributing to morbidity and death. The mechanisms by which unbalanced dietary nutrients affect intestinal homeostasis are largely unknown. Here we report that deficiency in murine angiotensin I converting enzyme (peptidyl-dipeptidase A) 2 (Ace2), which encodes a key regulatory enzyme of the renin-angiotensin system (RAS), results in highly increased susceptibility to intestinal inflammation induced by epithelial damage. The RAS is known to be involved in acute lung failure, cardiovascular functions and SARS infections. Mechanistically, ACE2 has a RAS-independent function, regulating intestinal amino acid homeostasis, expression of antimicrobial peptides, and the ecology of the gut microbiome. Transplantation of the altered microbiota from Ace2 mutant mice into germ-free wild-type hosts was able to transmit the increased propensity to develop severe colitis. ACE2-dependent changes in epithelial immunity and the gut microbiota can be directly regulated by the dietary amino acid tryptophan. Our results identify ACE2 as a key regulator of dietary amino acid homeostasis, innate immunity, gut microbial ecology, and transmissible susceptibility to colitis. These results provide a molecular explanation for how amino acid malnutrition can cause intestinal inflammation and diarrhoea.


Assuntos
Colite/etiologia , Colite/microbiologia , Intestinos/microbiologia , Desnutrição/complicações , Metagenoma , Peptidil Dipeptidase A/metabolismo , Triptofano/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Biocatálise , Colite/tratamento farmacológico , Colite/patologia , Sulfato de Dextrana , Diarreia/complicações , Proteínas Alimentares/metabolismo , Proteínas Alimentares/farmacologia , Feminino , Deleção de Genes , Predisposição Genética para Doença , Vida Livre de Germes , Homeostase , Imunidade Inata , Intestinos/patologia , Masculino , Desnutrição/metabolismo , Camundongos , Modelos Biológicos , Niacinamida/metabolismo , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Peptidil Dipeptidase A/deficiência , Peptidil Dipeptidase A/genética , Sistema Renina-Angiotensina/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Ácido Trinitrobenzenossulfônico , Triptofano/farmacologia , Triptofano/uso terapêutico
14.
Trends Endocrinol Metab ; 22(5): 188-94, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21470874

RESUMO

The receptor activator of nuclear factor-κB (RANK) and its ligand RANKL are best known for their essential function in bone remodeling and bone-related pathologies such as osteoporosis and arthritis. In humans, dysregulation of the RANK-RANKL system is the major cause of osteoporosis in postmenopausal women. Furthermore, appropriate RANKL signaling is also required for the formation of a lactating mammary gland. Both RANKL and RANK are expressed by mammary epithelial cells under the control of sex hormones. Recent data also indicate that RANK and RANKL control the preferential metastasis of breast cancer cells to the bone as well as sex hormone-driven primary mammary cancer. Here we critically review these data with special attention on mammary cancer development.


Assuntos
Neoplasias da Mama/etiologia , Hormônios Esteroides Gonadais/fisiologia , Metástase Neoplásica , Ligante RANK/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Animais , Neoplasias Ósseas/secundário , Remodelação Óssea/fisiologia , Mama/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Terapia de Reposição de Estrogênios/efeitos adversos , Feminino , Regulação da Expressão Gênica , Humanos , Lactação , Gravidez , Ligante RANK/antagonistas & inibidores , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética
15.
J Mol Med (Berl) ; 89(7): 647-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21445556

RESUMO

Receptor-activator of NF-κB ligand (TNFSF11, also known as RANKL, OPGL, TRANCE, and ODF) and its tumor necrosis factor (TNF)-family receptor RANK are essential regulators of bone remodeling, lymph node formation, establishment of the thymic microenvironment, mammary gland development during pregnancy, and bone metastasis in cancer. We have recently also reported that the RANKL/RANK system controls the incidence and onset of sex hormone, progestin-driven breast cancer. RANKL and RANK are also expressed in the central nervous systems where they play an essential role in body temperature regulation. RANKL activates brain regions involved in thermoregulation and induces fever via the COX2-PGE(2)/EP3R pathway. Moreover, female mice with a RANK gene deleted in neurons and astrocytes exhibit increased basal body temperature, suggesting that the RANKL/RANK system also controls physiological thermoregulation in females under the control of sex hormones. This review will summarize the recently emerging role of the RANKL/RANK signaling axis in mammary gland development, cancer metastasis, hormone-derived breast cancer development, and thermal regulation. Furthermore, we will highlight the striking therapeutic potential of this pathway and provide a molecular rationale for consideration of targeting RANKL/RANK in diseases such as breast cancer.


Assuntos
Osso e Ossos/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Lactação/metabolismo , Neoplasias/metabolismo , Ligante RANK/imunologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Transdução de Sinais/fisiologia
16.
Nat Genet ; 43(3): 212-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21317887

RESUMO

Most preneoplastic lesions are quiescent and do not progress to form overt tumors. It has been proposed that oncogenic stress activates the DNA damage response and the key tumor suppressor p53, which prohibits tumor growth. However, the molecular pathways by which cells sense a premalignant state in vivo are largely unknown. Here we report that tissue-specific inactivation of the stress signaling kinase MKK7 in KRas(G12D)-driven lung carcinomas and NeuT-driven mammary tumors markedly accelerates tumor onset and reduces overall survival. Mechanistically, MKK7 acts through the kinases JNK1 and JNK2, and this signaling pathway directly couples oncogenic and genotoxic stress to the stability of p53, which is required for cell cycle arrest and suppression of epithelial cancers. These results show that MKK7 functions as a major tumor suppressor in lung and mammary cancer in mouse and identify MKK7 as a vital molecular sensor to set a cellular anti-cancer barrier.


Assuntos
Dano ao DNA , Genes p53 , MAP Quinase Quinase 7/genética , Animais , Ciclo Celular/genética , Senescência Celular , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes ras , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Sistema de Sinalização das MAP Quinases , Neoplasias Mamárias Experimentais/genética , Camundongos , Estabilidade Proteica
17.
Nature ; 468(7320): 98-102, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20881962

RESUMO

Breast cancer is one of the most common cancers in humans and will on average affect up to one in eight women in their lifetime in the United States and Europe. The Women's Health Initiative and the Million Women Study have shown that hormone replacement therapy is associated with an increased risk of incident and fatal breast cancer. In particular, synthetic progesterone derivatives (progestins) such as medroxyprogesterone acetate (MPA), used in millions of women for hormone replacement therapy and contraceptives, markedly increase the risk of developing breast cancer. Here we show that the in vivo administration of MPA triggers massive induction of the key osteoclast differentiation factor RANKL (receptor activator of NF-κB ligand) in mammary-gland epithelial cells. Genetic inactivation of the RANKL receptor RANK in mammary-gland epithelial cells prevents MPA-induced epithelial proliferation, impairs expansion of the CD49f(hi) stem-cell-enriched population, and sensitizes these cells to DNA-damage-induced cell death. Deletion of RANK from the mammary epithelium results in a markedly decreased incidence and delayed onset of MPA-driven mammary cancer. These data show that the RANKL/RANK system controls the incidence and onset of progestin-driven breast cancer.


Assuntos
Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Progestinas/efeitos adversos , Ligante RANK/metabolismo , Animais , Apoptose/efeitos da radiação , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Feminino , Raios gama , Integrina alfa6/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Acetato de Medroxiprogesterona/administração & dosagem , Acetato de Medroxiprogesterona/efeitos adversos , Camundongos , NF-kappa B/metabolismo , Osteoclastos/citologia , Fosfoproteínas/análise , Fosfoproteínas/imunologia , Progestinas/administração & dosagem , Ligante RANK/deficiência , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA