Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Cell ; 12(5): 467-78, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17996650

RESUMO

Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disease (MPD) initiated by expression of the p210-BCR-ABL fusion protein. We demonstrate in a murine model of p210-BCR-ABL-induced MPD that gene targeting of Rac1 and Rac2 significantly delays or abrogates disease development. Attenuation of the disease phenotype is associated with severely diminished p210-BCR-ABL-induced downstream signaling in primary hematopoietic cells. We utilize NSC23766, a small molecule antagonist of Rac activation, to validate biochemically and functionally Rac as a molecular target in both a relevant animal model and in primary human CML cells in vitro and in a xenograft model in vivo, including in Imatinib-resistant p210-BCR-ABL disease. These data demonstrate that Rac is an additional therapeutic target in p210-BCR-ABL-mediated MPD.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Regulação Leucêmica da Expressão Gênica , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Proteínas rac de Ligação ao GTP/fisiologia , Aminoquinolinas/farmacologia , Animais , Antígenos CD34/biossíntese , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Camundongos , Transtornos Mieloproliferativos/terapia , Transplante de Neoplasias , Fenótipo , Pirimidinas/farmacologia , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína RAC2 de Ligação ao GTP
2.
Proc Natl Acad Sci U S A ; 109(8): 2772-7, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-21940501

RESUMO

Human breast cancers are broadly classified based on their gene-expression profiles into luminal- and basal-type tumors. These two major tumor subtypes express markers corresponding to the major differentiation states of epithelial cells in the breast: luminal (EpCAM(+)) and basal/myoepithelial (CD10(+)). However, there are also rare types of breast cancers, such as metaplastic carcinomas, where tumor cells exhibit features of alternate cell types that no longer resemble breast epithelium. Until now, it has been difficult to identify the cell type(s) in the human breast that gives rise to these various forms of breast cancer. Here we report that transformation of EpCAM(+) epithelial cells results in the formation of common forms of human breast cancer, including estrogen receptor-positive and estrogen receptor-negative tumors with luminal and basal-like characteristics, respectively, whereas transformation of CD10(+) cells results in the development of rare metaplastic tumors reminiscent of the claudin-low subtype. We also demonstrate the existence of CD10(+) breast cells with metaplastic traits that can give rise to skin and epidermal tissues. Furthermore, we show that the development of metaplastic breast cancer is attributable, in part, to the transformation of these metaplastic breast epithelial cells. These findings identify normal cellular precursors to human breast cancers and reveal the existence of a population of cells with epidermal progenitor activity within adult human breast tissues.


Assuntos
Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Adulto , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica/metabolismo , Molécula de Adesão da Célula Epitelial , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/metabolismo , Epitélio/patologia , Feminino , Humanos , Metaplasia , Neprilisina/metabolismo , Fenótipo
3.
Breast Cancer Res ; 16(5): 453, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25315014

RESUMO

INTRODUCTION: Lineage tracing studies in mice have revealed the localization and existence of lineage-restricted mammary epithelial progenitor cells that functionally contribute to expansive growth during puberty and differentiation during pregnancy. However, extensive anatomical differences between mouse and human mammary tissues preclude the direct translation of rodent findings to the human breast. Therefore, here we characterize the mammary progenitor cell hierarchy and identify the anatomic location of progenitor cells within human breast tissues. METHODS: Mammary epithelial cells (MECs) were isolated from disease-free reduction mammoplasty tissues and assayed for stem/progenitor activity in vitro and in vivo. MECs were sorted and evaluated for growth on collagen and expression of lineages markers. Breast lobules were microdissected and individually characterized based on lineage markers and steroid receptor expression to identify the anatomic location of progenitor cells. Spanning-tree progression analysis of density-normalized events (SPADE) was used to identify the cellular hierarchy of MECs within lobules from high-dimensional cytometry data. RESULTS: Integrating multiple assays for progenitor activity, we identified the presence of luminal alveolar and basal ductal progenitors. Further, we show that Type I lobules of the human breast were the least mature, demonstrating an unrestricted pattern of expression of luminal and basal lineage markers. Consistent with this, SPADE analysis revealed that immature lobules were enriched for basal progenitor cells, while mature lobules consisted of increased hierarchal complexity of cells within the luminal lineages. CONCLUSIONS: These results reveal underlying differences in the human breast epithelial hierarchy and suggest that with increasing glandular maturity, the epithelial hierarchy also becomes more complex.


Assuntos
Células-Tronco Adultas/fisiologia , Glândulas Mamárias Humanas/citologia , Células-Tronco Adultas/transplante , Animais , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Queratina-14/metabolismo , Queratina-18/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID
4.
Cancer Res ; 84(15): 2501-2517, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38833522

RESUMO

Recurrent somatic mutations in the BRG1/BRM-associated factor (BAF) chromatin remodeling complex subunit ARID1A occur frequently in advanced urothelial, endometrial, and ovarian clear cell carcinomas, creating an alternative chromatin state that may be exploited therapeutically. The histone methyltransferase EZH2 has been previously identified as targetable vulnerability in the context of ARID1A mutations. In this study, we describe the discovery of tulmimetostat, an orally available, clinical stage EZH2 inhibitor, and it elucidates the aspects of its application potential in ARID1A mutant tumors. Tulmimetostat administration achieved efficacy in multiple ARID1A mutant bladder, ovarian, and endometrial tumor models and improved cisplatin response in chemotherapy-resistant models. Consistent with its comprehensive and durable level of target coverage, tulmimetostat demonstrated greater efficacy than other PRC2-targeted inhibitors at comparable or lower exposures in a bladder cancer xenograft mouse model. Tulmimetostat mediated extensive changes in gene expression, in addition to a profound reduction in global H3K27me3 levels in tumors. Phase I clinical pharmacokinetic and pharmacodynamic data indicated that tulmimetostat exhibits durable exposure and profound target engagement. Importantly, a tulmimetostat controlled gene expression signature identified in whole blood from a cohort of 32 patients with cancer correlated with tulmimetostat exposure, representing a pharmacodynamic marker for the assessment of target coverage for PRC2-targeted agents in the clinic. Collectively, these data suggest that tulmimetostat has the potential to achieve clinical benefit in solid tumors as a monotherapy but also in combination with chemotherapeutic agents, and may be beneficial in various indications with recurrent ARID1A mutations. Significance: The EZH2 inhibitor tulmimetostat achieves comprehensive target inhibition in ARID1A mutant solid tumor models and cancer patients that can be assessed with a pharmacodynamic gene signature in peripheral blood.


Assuntos
Proteínas de Ligação a DNA , Proteína Potenciadora do Homólogo 2 de Zeste , Mutação , Fatores de Transcrição , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Feminino , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Linhagem Celular Tumoral , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo
5.
Proc Natl Acad Sci U S A ; 107(50): 21737-42, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21098263

RESUMO

Many tumors contain heterogeneous populations of cells, only some of which exhibit increased tumorigenicity and resistance to anticancer therapies. Evidence suggests that these aggressive cancer cells, often termed "cancer stem cells" or "cancer stem-like cells" (CSCs), rely upon developmental signaling pathways that are important for survival and expansion of normal stem cells. Here we report that, in analogy to embryonic mammary epithelial biology, estrogen signaling expands the pool of functional breast CSCs through a paracrine FGF/FGFR/Tbx3 signaling pathway. Estrogen or FGF9 pretreatment induced CSC properties of breast cancer cell lines and freshly isolated breast cancer cells, whereas cotreatment of cells with tamoxifen or a small molecule inhibitor of FGFR signaling was sufficient to prevent the estrogen-induced expansion of CSCs. Furthermore, reduction of FGFR or Tbx3 gene expression was able to abrogate tumorsphere formation, whereas ectopic Tbx3 expression increased tumor seeding potential by 100-fold. These findings demonstrate that breast CSCs are stimulated by estrogen through a signaling pathway that similarly controls normal mammary epithelial stem cell biology.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Comunicação Parácrina , Transdução de Sinais/efeitos dos fármacos , Proteínas com Domínio T/metabolismo , Linhagem Celular Tumoral , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Humanos , Células-Tronco Neoplásicas/fisiologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas com Domínio T/genética
6.
Semin Cell Dev Biol ; 21(1): 11-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19857593

RESUMO

It is increasingly apparent that normal and malignant breast tissues require complex local and systemic stromal interactions for development and progression. During development, mammary cell fate specification and differentiation require highly regulated contextual signals derived from the stroma. Likewise, during breast carcinoma development, the tissue stroma can provide tumor suppressing and tumor-promoting environments that serve to regulate neoplastic growth of the epithelium. This review focuses on the role of the stroma as a mediator of normal mammary development, as well as a critical regulator of malignant conversion and progression in breast cancer. Recognition of the important role of the stroma during the progression of breast cancers leads to the possibility of new targets for treatment of the initial breast cancer lesion as well as prevention of recurrence.


Assuntos
Neoplasias da Mama , Mama , Tecido Conjuntivo , Células Estromais/fisiologia , Animais , Mama/anatomia & histologia , Mama/crescimento & desenvolvimento , Mama/patologia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Transformação Celular Neoplásica , Tecido Conjuntivo/anatomia & histologia , Tecido Conjuntivo/patologia , Progressão da Doença , Feminino , Fibrose/patologia , Humanos , Células Estromais/citologia , Células Estromais/patologia
7.
Biochem J ; 424(1): 153-61, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19723022

RESUMO

The Rnd proteins (Rnd1, Rnd2 and Rnd3/RhoE) form a distinct branch of the Rho family of small GTPases. Altered Rnd3 expression causes changes in cytoskeletal organization and cell cycle progression. Rnd3 functions to decrease RhoA activity, but how Rnd3 itself is regulated to cause these changes is still under investigation. Unlike other Rho family proteins, Rnd3 is regulated not by GTP/GDP cycling, but at the level of expression and by post-translational modifications such as prenylation and phosphorylation. We show in the present study that, upon PKC (protein kinase C) agonist stimulation, Rnd3 undergoes an electrophoretic mobility shift and its subcellular localization becomes enriched at internal membranes. These changes are blocked by inhibition of conventional PKC isoforms and do not occur in PKCalpha-null cells or to a non-phosphorylatable mutant of Rnd3. We further show that PKCalpha directly phosphorylates Rnd3 in an in vitro kinase assay. Additionally, we provide evidence that the phosphorylation status of Rnd3 has a direct effect on its ability to block signalling from the Rho-ROCK (Rho-kinase) pathway. These results identify an additional mechanism of regulation and provide clarification of how Rnd3 modulates Rho signalling to alter cytoskeletal organization.


Assuntos
Proteína Quinase C-alfa/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Western Blotting , Humanos , Camundongos , Células NIH 3T3 , Fosforilação/genética , Proteína Quinase C-alfa/genética , Ratos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/genética
8.
Cancer Res ; 66(6): 3153-61, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540666

RESUMO

Phosphatase found in regenerating liver (PRL)-1, PRL-2, and PRL-3 [also known as PTP4A1, PTP4A2, and PTP4A3, respectively] constitute a unique family of putative protein tyrosine phosphatases (PTPs) modified by farnesylation. PRL-3 is amplified and its message is up-regulated in colorectal carcinoma metastases. Its ectopic expression promotes invasive and metastatic properties, supporting a causal link between PRL-3 and late-stage cancer development. However, neither PRL phosphatase substrates nor their signaling pathways have been defined. To address possible mechanisms for the biological activity of PRL-3, we sought to identify its downstream targets, reasoning that regulators of motility and invasion, such as the Rho family of small GTPases, might be logical candidates. We found that levels of active RhoA and RhoC were increased 4- to 7-fold in SW480 colorectal carcinoma cells expressing exogenous PRL-1 and PRL-3, and that PRL-mediated motility and Matrigel invasion were blocked by pharmacologic inhibition of Rho kinase (ROCK), a key Rho effector. In contrast, the activity of Rac was reduced by PRL PTPs, whereas Cdc42 activity was unaffected. PRL-3 stimulated transcription driven by the serum response element in a Rho-dependent manner. We also confirmed that the ability of PRL PTPs to induce invasion and motility is dependent on farnesylation. Catalytic PRL-3 mutants (C104A or D72A) were impaired in PRL-3-induced invasion and Rho activation, indicating that these properties require phosphatase activity. We conclude that PRL PTPs stimulate Rho signaling pathways to promote motility and invasion. Characterization of PRL activity and regulatory pathways should enhance efforts to understand and interfere with PRL-mediated events in invasion and metastasis.


Assuntos
Adenocarcinoma/enzimologia , Proteínas de Ciclo Celular/metabolismo , Movimento Celular/fisiologia , Neoplasias do Colo/enzimologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas de Membrana , Camundongos , Células NIH 3T3 , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Prenilação de Proteína , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transfecção , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas ras , Proteína de Ligação a GTP rhoC
9.
Cancer Res ; 65(21): 9883-90, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16267012

RESUMO

Our initial characterization of Rac3, a close relative of the small GTPase Rac1, established its ability to promote membrane ruffling, transformation, and activation of c-jun transcriptional activity. The finding that Rac3 is transforming, and its similarity to Rac1, a protein that has a well-established connection to many processes important for cancer progression, prompted further investigation into Rac3 transformation. We used effector domain mutants (EDMs) to explore the relationship among Rac signaling, transformation, and effector usage. All Rac3 EDMs tested (N26D, F37L, Y40C, and N43D) retained the ability to promote membrane ruffling and focus formation. In contrast, only the N43D mutant promoted anchorage independence. This differs from Rac1, where both N26D and N43D mutants were impaired in both types of transformation. To learn more about the signaling pathways involved, we did luciferase reporter assays and glutathione S-transferase pull-down assays for effector binding. We found evidence for a functional link between activation of phospholipase Cbeta2 by Rac3 and signaling to the serum response factor (SRF). Surprisingly, we also found that Rac3 binds poorly to the known Rac1 effectors mixed lineage kinases 2 and 3 (MLK2 and MLK3). Transcription of cyclin D1 was the only pathway that correlated with growth in soft agar. Our experiments show that activation of membrane ruffling and transcriptional activation of c-jun, SRF, or E2F are not sufficient to promote anchorage-independent growth mediated by Rac3. Instead, multiple effector pathways are required for Rac3 transformation, and these overlap partially but not completely with those used by Rac1.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas rac de Ligação ao GTP/genética , Animais , Células COS , Proteínas de Transporte/metabolismo , Adesão Celular/genética , Processos de Crescimento Celular/genética , Transformação Celular Neoplásica/patologia , Chlorocebus aethiops , Isoenzimas/metabolismo , Camundongos , Células NIH 3T3 , Neuropeptídeos/metabolismo , Fosfolipase C beta , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Fator de Resposta Sérica/biossíntese , Fator de Resposta Sérica/genética , Ativação Transcricional , Fosfolipases Tipo C/metabolismo , Quinases Ativadas por p21 , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
10.
Stem Cell Reports ; 9(3): 943-955, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28781076

RESUMO

Organogenesis and tissue development occur through sequential stepwise processes leading to increased lineage restriction and loss of pluripotency. An exception to this appears in the adult human breast, where rare variant epithelial cells exhibit pluripotency and multilineage differentiation potential when removed from the signals of their native microenvironment. This phenomenon provides a unique opportunity to study mechanisms that lead to cellular reprogramming and lineage plasticity in real time. Here, we show that primary human mammary epithelial cells (HMECs) lose expression of differentiated mammary epithelial markers in a manner dependent on paracrine factors and epigenetic regulation. Furthermore, we demonstrate that HMEC reprogramming is dependent on gene silencing by the DNA methyltransferase DNMT3A and loss of histone transcriptional marks following downregulation of the methyltransferase DOT1L. These results demonstrate that lineage commitment in adult tissues is context dependent and highlight the plasticity of somatic cells when removed from their native tissue microenvironment.


Assuntos
Linhagem da Célula/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética , Células Epiteliais/metabolismo , Glândulas Mamárias Humanas/citologia , Metiltransferases/metabolismo , Diferenciação Celular/genética , Microambiente Celular , Reprogramação Celular/genética , DNA Metiltransferase 3A , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Metilação , Células Estromais/citologia
11.
Elife ; 52016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26731516

RESUMO

Pharmacological inhibition of chromatin co-regulatory factors represents a clinically validated strategy to modulate oncogenic signaling through selective attenuation of gene expression. Here, we demonstrate that CBP/EP300 bromodomain inhibition preferentially abrogates the viability of multiple myeloma cell lines. Selective targeting of multiple myeloma cell lines through CBP/EP300 bromodomain inhibition is the result of direct transcriptional suppression of the lymphocyte-specific transcription factor IRF4, which is essential for the viability of myeloma cells, and the concomitant repression of the IRF4 target gene c-MYC. Ectopic expression of either IRF4 or MYC antagonizes the phenotypic and transcriptional effects of CBP/EP300 bromodomain inhibition, highlighting the IRF4/MYC axis as a key component of its mechanism of action. These findings suggest that CBP/EP300 bromodomain inhibition represents a viable therapeutic strategy for targeting multiple myeloma and other lymphoid malignancies dependent on the IRF4 network.


Assuntos
Antineoplásicos/farmacologia , Proteína p300 Associada a E1A/antagonistas & inibidores , Fatores Reguladores de Interferon/metabolismo , Mieloma Múltiplo/fisiopatologia , Fragmentos de Peptídeos/antagonistas & inibidores , Sialoglicoproteínas/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos
12.
ACS Med Chem Lett ; 7(5): 531-6, 2016 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-27190605

RESUMO

CBP and EP300 are highly homologous, bromodomain-containing transcription coactivators involved in numerous cellular pathways relevant to oncology. As part of our effort to explore the potential therapeutic implications of selectively targeting bromodomains, we set out to identify a CBP/EP300 bromodomain inhibitor that was potent both in vitro and in cellular target engagement assays and was selective over the other members of the bromodomain family. Reported here is a series of cell-potent and selective probes of the CBP/EP300 bromodomains, derived from the fragment screening hit 4-methyl-1,3,4,5-tetrahydro-2H-benzo[b][1,4]diazepin-2-one.

13.
Nat Commun ; 6: 7505, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26106036

RESUMO

Although BRCA1 function is essential for maintaining genomic integrity in all cell types, it is unclear why increased risk of cancer in individuals harbouring deleterious mutations in BRCA1 is restricted to only a select few tissues. Here we show that human mammary epithelial cells (HMECs) from BRCA1-mutation carriers (BRCA1(mut/+)) exhibit increased genomic instability and rapid telomere erosion in the absence of tumour-suppressor loss. Furthermore, we uncover a novel form of haploinsufficiency-induced senescence (HIS) specific to epithelial cells, which is triggered by pRb pathway activation rather than p53 induction. HIS and telomere erosion in HMECs correlate with misregulation of SIRT1 leading to increased levels of acetylated pRb as well as acetylated H4K16 both globally and at telomeric regions. These results identify a novel form of cellular senescence and provide a potential molecular basis for the rapid cell- and tissue- specific predisposition of breast cancer development associated with BRCA1 haploinsufficiency.


Assuntos
Senescência Celular/genética , Células Epiteliais/metabolismo , Genes BRCA1 , Instabilidade Genômica/genética , Haploinsuficiência , Glândulas Mamárias Humanas/metabolismo , Encurtamento do Telômero/genética , Dano ao DNA , Células Epiteliais/citologia , Heterozigoto , Humanos , Glândulas Mamárias Humanas/citologia , Mutação , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
14.
Cancer Res ; 73(19): 6080-93, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23959857

RESUMO

Obesity is one of the most important preventable causes of cancer and the most significant risk factor for breast cancer in postmenopausal women. Compared with lean women, obese women are more likely to be diagnosed with a larger, higher grade tumor, an increased incidence of lymph node metastases, and elevated risk of distant recurrence. However, the mechanisms connecting obesity to the pathogenesis of breast cancer are poorly defined. Here, we show that during obesity, adipocytes within human and mouse breast tissues recruit and activate macrophages through a previously uncharacterized CCL2/IL-1ß/CXCL12 signaling pathway. Activated macrophages in turn promote stromal vascularization and angiogenesis even before the formation of cancer. Recapitulating these changes using a novel humanized breast cancer model was sufficient to promote angiogenesis and prime the microenvironment prior to neoplastic transformation for accelerated breast oncogenesis. These findings provide a mechanistic role for adipocytes and macrophages before carcinogenesis that may be critical for prevention and treatment of obesity-related cancer.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Quimiocina CCL2/metabolismo , Macrófagos/patologia , Neovascularização Patológica , Obesidade/complicações , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Apoptose , Western Blotting , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Bovinos , Diferenciação Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CXCL12/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Obesidade/metabolismo , Obesidade/patologia , Retina/citologia , Retina/metabolismo , Microambiente Tumoral
15.
Cell Stem Cell ; 9(6): 496-7, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-22136921

RESUMO

Transplantation assays suggest that multipotent stem cells maintain the two lineages of the mammary gland. Recently in Nature, Van Keymeulen et al. (2011) used lineage tracing to discover unipotent stem cells that maintain the bulk of the mouse mammary gland after birth and during pregnancy.

16.
Cell Stem Cell ; 8(2): 149-63, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21295272

RESUMO

Women with inherited mutations in the BRCA1 gene have increased risk of developing breast cancer but also exhibit a predisposition for the development of aggressive basal-like breast tumors. We report here that breast epithelial cells derived from patients harboring deleterious mutations in BRCA1 (BRCA1(mut /+) give rise to tumors with increased basal differentiation relative to cells from BRCA1+/+ patients. Molecular analysis of disease-free breast tissues from BRCA1(mut /+) patients revealed defects in progenitor cell lineage commitment even before cancer incidence. Moreover, we discovered that the transcriptional repressor Slug is an important functional suppressor of human breast progenitor cell lineage commitment and differentiation and that it is aberrantly expressed in BRCA1(mut /+) tissues. Slug expression is necessary for increased basal-like phenotypes prior to and after neoplastic transformation. These findings demonstrate that the genetic background of patient populations, in addition to affecting incidence rates, significantly impacts progenitor cell fate commitment and, therefore, tumor phenotype.


Assuntos
Neoplasias da Mama/metabolismo , Predisposição Genética para Doença/genética , Células-Tronco/citologia , Adulto , Animais , Neoplasias da Mama/genética , Feminino , Citometria de Fluxo , Genes BRCA1 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
J Biol Chem ; 283(37): 25150-25163, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18614539

RESUMO

Rho GTPases (20 human members) comprise a major branch of the Ras superfamily of small GTPases, and aberrant Rho GTPase function has been implicated in oncogenesis and other human diseases. Although many of our current concepts of Rho GTPases are based on the three classical members (RhoA, Rac1, and Cdc42), recent studies have revealed the diversity of biological functions mediated by other family members. A key basis for the functional diversity of Rho GTPases is their association with distinct subcellular compartments, which is dictated in part by three posttranslational modifications signaled by their carboxyl-terminal CAAX (where C represents cysteine, A is an aliphatic amino acid, and X is a terminal amino acid) tetrapeptide motifs. CAAX motifs are substrates for the prenyltransferase-catalyzed addition of either farnesyl or geranylgeranyl isoprenoid lipids, Rce1-catalyzed endoproteolytic cleavage of the AAX amino acids, and Icmt-catalyzed carboxyl methylation of the isoprenylcysteine. We utilized pharmacologic, biochemical, and genetic approaches to determine the sequence requirements and roles of CAAX signal modifications in dictating the subcellular locations and functions of the Rho GTPase family. Although the classical Rho GTPases are modified by geranylgeranylation, we found that a majority of the other Rho GTPases are substrates for farnesyltransferase. We found that the membrane association and/or function of Rho GTPases are differentially dependent on Rce1- and Icmt-mediated modifications. Our results further delineate the sequence requirements for prenyltransferase specificity and functional roles for protein prenylation in Rho GTPase function. We conclude that a majority of Rho GTPases are targets for pharmacologic inhibitors of farnesyltransferase, Rce1, and Icmt.


Assuntos
Proteínas rho de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Cisteína/química , Endopeptidases/química , Farnesiltranstransferase/antagonistas & inibidores , Humanos , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
19.
Methods ; 37(2): 131-37, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16288891

RESUMO

Modification of small GTPases by lipids is required for their proper subcellular localization and biological activity. Lipids added post-translationally include both farnesyl and geranylgeranyl isoprenoids and the fatty acid palmitate. Thus, specific small molecule inhibitors of these processes cause mislocalization of small GTPases and impair their biological activity. Common biochemical methods of determining the lipid modification status or inhibitor sensitivity of small GTPases, such as in vitro prenylation assays, SDS-PAGE mobility shifts or metabolic labeling, although highly useful in their own right, cannot distinguish differences among specific subpopulations of cells, link lipid modification status with other properties of interest, or provide spatio-temporal information. An alternative method takes advantage of the tight link between small GTPase lipid modification and subcellular localization. The innate localization pattern of the enhanced green fluorescent protein, a common epitope tag frequently used in live cell imaging, is altered by fusion to modified but not unmodified small GTPases. We describe here a technique that takes advantage of these properties to monitor post-translational modifications of these proteins in a rapid, visual manner in live cells.


Assuntos
GTP Fosfo-Hidrolases/genética , Proteínas de Fluorescência Verde/genética , Metabolismo dos Lipídeos , Sondas Moleculares , Processamento de Proteína Pós-Traducional/fisiologia , Animais , GTP Fosfo-Hidrolases/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Células NIH 3T3 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA