Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(41): E9640-E9648, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30242135

RESUMO

Breast cancer stem cells (BCSCs), which are characterized by a capacity for unlimited self-renewal and for generation of the bulk cancer cell population, play a critical role in cancer relapse and metastasis. Hypoxia is a common feature of the cancer microenvironment that stimulates the specification and maintenance of BCSCs. In this study, we found that hypoxia increased expression of adenosine receptor 2B (A2BR) in human breast cancer cells through the transcriptional activity of hypoxia-inducible factor 1. The binding of adenosine to A2BR promoted BCSC enrichment by activating protein kinase C-δ, which phosphorylated and activated the transcription factor STAT3, leading to increased expression of interleukin 6 and NANOG, two key mediators of the BCSC phenotype. Genetic or pharmacological inhibition of A2BR expression or activity decreased hypoxia- or adenosine-induced BCSC enrichment in vitro, and dramatically impaired tumor initiation and lung metastasis after implantation of MDA-MB-231 human breast cancer cells into the mammary fat pad of immunodeficient mice. These data provide evidence that targeting A2BR might be an effective strategy to eradicate BCSCs.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor A2B de Adenosina/biossíntese , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Fator 1 Induzível por Hipóxia/genética , Células MCF-7 , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Receptor A2B de Adenosina/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(6): E1239-E1248, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29367423

RESUMO

Triple-negative breast cancer (TNBC) is treated with cytotoxic chemotherapy and is often characterized by early relapse and metastasis. To form a secondary (recurrent and/or metastatic) tumor, a breast cancer cell must evade the innate and adaptive immune systems. CD47 enables cancer cells to evade killing by macrophages, whereas CD73 and PDL1 mediate independent mechanisms of evasion of cytotoxic T lymphocytes. Here, we report that treatment of human or murine TNBC cells with carboplatin, doxorubicin, gemcitabine, or paclitaxel induces the coordinate transcriptional induction of CD47, CD73, and PDL1 mRNA and protein expression, leading to a marked increase in the percentage of CD47+CD73+PDL1+ breast cancer cells. Genetic or pharmacological inhibition of hypoxia-inducible factors (HIFs) blocked chemotherapy-induced enrichment of CD47+CD73+PDL1+ TNBC cells, which were also enriched in the absence of chemotherapy by incubation under hypoxic conditions, leading to T cell anergy or death. Treatment of mice with cytotoxic chemotherapy markedly increased the intratumoral ratio of regulatory/effector T cells, an effect that was abrogated by HIF inhibition. Our results delineate an HIF-dependent transcriptional mechanism contributing to TNBC progression and suggest that combining chemotherapy with an HIF inhibitor may prevent countertherapeutic induction of proteins that mediate evasion of innate and adaptive antitumor immunity.


Assuntos
5'-Nucleotidase/metabolismo , Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Antígeno CD47/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias de Mama Triplo Negativas/imunologia , Evasão Tumoral/imunologia , 5'-Nucleotidase/genética , Animais , Apoptose , Antígeno B7-H1/genética , Antígeno CD47/genética , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 113(14): E2047-56, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27001847

RESUMO

N(6)-methyladenosine (m(6)A) modification of mRNA plays a role in regulating embryonic stem cell pluripotency. However, the physiological signals that determine the balance between methylation and demethylation have not been described, nor have studies addressed the role of m(6)A in cancer stem cells. We report that exposure of breast cancer cells to hypoxia stimulated hypoxia-inducible factor (HIF)-1α- and HIF-2α-dependent expression of AlkB homolog 5 (ALKBH5), an m(6)A demethylase, which demethylated NANOG mRNA, which encodes a pluripotency factor, at an m(6)A residue in the 3'-UTR. Increased NANOG mRNA and protein expression, and the breast cancer stem cell (BCSC) phenotype, were induced by hypoxia in an HIF- and ALKBH5-dependent manner. Insertion of the NANOG 3'-UTR into a luciferase reporter gene led to regulation of luciferase activity by O2, HIFs, and ALKBH5, which was lost upon mutation of the methylated residue. ALKBH5 overexpression decreased NANOG mRNA methylation, increased NANOG levels, and increased the percentage of BCSCs, phenocopying the effect of hypoxia. Knockdown of ALKBH5 expression in MDA-MB-231 human breast cancer cells significantly reduced their capacity for tumor initiation as a result of reduced numbers of BCSCs. Thus, HIF-dependent ALKBH5 expression mediates enrichment of BCSCs in the hypoxic tumor microenvironment.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Neoplasias da Mama/patologia , Hipóxia Celular , Proteína Homeobox Nanog/genética , Células-Tronco Neoplásicas/patologia , RNA Mensageiro/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Catálise , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Metilação
4.
Proc Natl Acad Sci U S A ; 112(33): E4600-9, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26229077

RESUMO

Triple negative breast cancer (TNBC) accounts for 10-15% of all breast cancer but is responsible for a disproportionate share of morbidity and mortality because of its aggressive characteristics and lack of targeted therapies. Chemotherapy induces enrichment of breast cancer stem cells (BCSCs), which are responsible for tumor recurrence and metastasis. Here, we demonstrate that chemotherapy induces the expression of the cystine transporter xCT and the regulatory subunit of glutamate-cysteine ligase (GCLM) in a hypoxia-inducible factor (HIF)-1-dependent manner, leading to increased intracellular glutathione levels, which inhibit mitogen-activated protein kinase kinase (MEK) activity through copper chelation. Loss of MEK-ERK signaling causes FoxO3 nuclear translocation and transcriptional activation of the gene encoding the pluripotency factor Nanog, which is required for enrichment of BCSCs. Inhibition of xCT, GCLM, FoxO3, or Nanog blocks chemotherapy-induced enrichment of BCSCs and impairs tumor initiation. These results suggest that, in combination with chemotherapy, targeting BCSCs by inhibiting HIF-1-regulated glutathione synthesis may improve outcome in TNBC.


Assuntos
Antineoplásicos/química , Cobre/química , Glutationa/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/citologia , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Linhagem Celular Tumoral , Quelantes/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos SCID , Transplante de Neoplasias , Oligonucleotídeos/genética , Paclitaxel/química , Fenótipo , Fosforilação , RNA Mensageiro/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(45): E6215-23, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26512116

RESUMO

Increased expression of CD47 has been reported to enable cancer cells to evade phagocytosis by macrophages and to promote the cancer stem cell phenotype, but the molecular mechanisms regulating CD47 expression have not been determined. Here we report that hypoxia-inducible factor 1 (HIF-1) directly activates transcription of the CD47 gene in hypoxic breast cancer cells. Knockdown of HIF activity or CD47 expression increased the phagocytosis of breast cancer cells by bone marrow-derived macrophages. CD47 expression was increased in mammosphere cultures, which are enriched for cancer stem cells, and CD47 deficiency led to cancer stem cell depletion. Analysis of datasets derived from thousands of patients with breast cancer revealed that CD47 expression was correlated with HIF target gene expression and with patient mortality. Thus, CD47 expression contributes to the lethal breast cancer phenotype that is mediated by HIF-1.


Assuntos
Neoplasias da Mama/metabolismo , Antígeno CD47/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/fisiologia , Fagocitose/fisiologia , Evasão Tumoral/fisiologia , Análise de Variância , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fator 1 Induzível por Hipóxia/farmacologia , Immunoblotting , Luciferases , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real
6.
Proc Natl Acad Sci U S A ; 111(50): E5429-38, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25453096

RESUMO

Triple negative breast cancers (TNBCs) are defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 expression, and are treated with cytotoxic chemotherapy such as paclitaxel or gemcitabine, with a durable response rate of less than 20%. TNBCs are enriched for the basal subtype gene expression profile and the presence of breast cancer stem cells, which are endowed with self-renewing and tumor-initiating properties and resistance to chemotherapy. Hypoxia-inducible factors (HIFs) and their target gene products are highly active in TNBCs. Here, we demonstrate that HIF expression and transcriptional activity are induced by treatment of MDA-MB-231, SUM-149, and SUM-159, which are human TNBC cell lines, as well as MCF-7, which is an ER(+)/PR(+) breast cancer line, with paclitaxel or gemcitabine. Chemotherapy-induced HIF activity enriched the breast cancer stem cell population through interleukin-6 and interleukin-8 signaling and increased expression of multidrug resistance 1. Coadministration of HIF inhibitors overcame the resistance of breast cancer stem cells to paclitaxel or gemcitabine, both in vitro and in vivo, leading to tumor eradication. Increased expression of HIF-1α or HIF target genes in breast cancer biopsies was associated with decreased overall survival, particularly in patients with basal subtype tumors and those treated with chemotherapy alone. Based on these results, clinical trials are warranted to test whether treatment of patients with TNBC with a combination of cytotoxic chemotherapy and HIF inhibitors will improve patient survival.


Assuntos
Resistencia a Medicamentos Antineoplásicos/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Análise de Variância , Linhagem Celular Tumoral , Primers do DNA/genética , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Processamento de Imagem Assistida por Computador , Immunoblotting , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Estimativa de Kaplan-Meier , Luciferases , Microscopia , Paclitaxel/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/metabolismo , Gencitabina
7.
Cell Rep ; 32(8): 108073, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32846130

RESUMO

Immune checkpoint blockade (ICB) has led to therapeutic responses in some cancer patients for whom no effective treatment previously existed. ICB acts on T lymphocytes and other immune cells that are inactivated due to checkpoint signals that inhibit their infiltration and function within tumors. But for more than 80% of patients, immunotherapy has not been effective. Here, we demonstrate a cancer-cell-intrinsic mechanism of immune evasion and resistance to ICB mediated by baculoviral IAP repeat-containing 2 (BIRC2). Knockdown of BIRC2 expression in mouse melanoma or breast cancer cells increases expression of the chemokine CXCL9 and impairs tumor growth by increasing the number of intratumoral activated CD8+ T cells and natural killer cells. Administration of anti-CXCL9 neutralizing antibody inhibits the recruitment of CD8+ T cells and natural killer cells to BIRC2-deficient tumors. Most importantly, BIRC2 deficiency dramatically increases the sensitivity of mouse melanoma and breast tumors to anti-CTLA4 and/or anti-PD1 ICB.


Assuntos
Imunidade Inata/imunologia , Imunoterapia/métodos , Proteínas Inibidoras de Apoptose/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Humanos , Camundongos , Microambiente Tumoral
9.
Methods Mol Biol ; 1742: 237-246, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29330805

RESUMO

Aldehyde dehydrogenase and mammosphere assays enable the cost-effective quantification and characterization of cancer stem cells (CSCs) from cancer cell lines as well as cancer tissue. Here we describe the quantification of CSCs in breast cancer cell lines using aldehyde dehydrogenase and mammosphere assays under hypoxic (1% O2) and non-hypoxic (20% O2) culture conditions. Using this method, a significant enrichment of CSCs compared to bulk populations is observed when breast cancer cells are exposed to 1% O2 for 72 h.


Assuntos
Aldeído Desidrogenase/metabolismo , Neoplasias da Mama/patologia , Células-Tronco Neoplásicas/citologia , Neoplasias da Mama/metabolismo , Hipóxia Celular , Feminino , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Células MCF-7 , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Esferoides Celulares , Células Tumorais Cultivadas
10.
Biochim Biophys Acta Rev Cancer ; 1870(1): 15-22, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30006019

RESUMO

Cancer cells are characterized by high metabolic demand. The substrates in demand include oxygen, glucose, glutamine and lipids. Oxygen serves as a key substrate in cellular metabolism and bioenergetics. Hypoxia or low oxygen abundance is a common feature of the tumor microenvironment that occurs due to an imbalance in supply and demand. Many of the metabolic responses to hypoxia in both cancer cells and stromal cells are orchestrated by hypoxia-inducible factors (HIFs). In this review we summarize our current understanding of how HIFs modulate the metabolism of hypoxic cancer cells and immune cells, and how altered metabolism plays a role in cancer progression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias/metabolismo , Adenosina/metabolismo , Aminoácidos/metabolismo , Translocador Nuclear Receptor Aril Hidrocarboneto , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Hipóxia Celular , Ácidos Graxos/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Humanos , Lipídeos/biossíntese , Neoplasias/tratamento farmacológico , Linfócitos T/metabolismo
11.
Redox Biol ; 13: 331-335, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28624704

RESUMO

Oxidative phosphorylation enables cells to generate the large amounts of ATP required for development and maintenance of multicellular organisms. However, under conditions of reduced O2 availability, electron transport becomes less efficient, leading to increased generation of superoxide anions. Hypoxia-inducible factors switch cells from oxidative to glycolytic metabolism, to reduce mitochondrial superoxide generation, and increase the synthesis of NADPH and glutathione, in order to maintain redox homeostasis under hypoxic conditions.


Assuntos
Homeostase , Fator 1 Induzível por Hipóxia/metabolismo , Estresse Oxidativo , Acetilcoenzima A/metabolismo , Animais , Humanos , Fator 1 Induzível por Hipóxia/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
12.
Artigo em Inglês | MEDLINE | ID: mdl-28221004

RESUMO

Metazoan species maintain oxygen homeostasis through the activity of hypoxia-inducible factors, which are transcriptional activators that regulate the expression of hundreds of genes to match O2 supply and demand. Here, we review the involvement of hypoxia-inducible factors in the molecular physiology and pathophysiology of cellular O2 sensing, O2 delivery, O2 utilization, and systemic O2 sensing. WIREs Syst Biol Med 2017, 9:e1382. doi: 10.1002/wsbm.1382 For further resources related to this article, please visit the WIREs website.


Assuntos
Homeostase/fisiologia , Modelos Cardiovasculares , Oxigênio/metabolismo , Biologia de Sistemas , Humanos
14.
Mol Cancer Res ; 15(6): 723-734, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28213554

RESUMO

Metastasis is the leading cause of breast cancer mortality. Previous studies have implicated hypoxia-induced changes in the composition and stiffness of the extracellular matrix (ECM) in the metastatic process. Therefore, the contribution of potential ECM-binding receptors in this process was explored. Using a bioinformatics approach, the expression of all integrin receptor subunits, in two independent breast cancer patient datasets, were analyzed to determine whether integrin status correlates with a validated hypoxia-inducible gene signature. Subsequently, a large panel of breast cancer cell lines was used to validate that hypoxia induces the expression of integrins that bind to collagen (ITGA1, ITGA11, ITGB1) and fibronectin (ITGA5, ITGB1). Hypoxia-inducible factors (HIF-1 and HIF-2) are directly required for ITGA5 induction under hypoxic conditions, which leads to enhanced migration and invasion of single cells within a multicellular 3D tumor spheroid but did not affect migration in a 2D microenvironment. ITGB1 expression requires HIF-1α, but not HIF-2α, for hypoxic induction in breast cancer cells. ITGA5 (α5 subunit) is required for metastasis to lymph nodes and lungs in breast cancer models, and high ITGA5 expression in clinical biopsies is associated with an increased risk of mortality.Implications: These results reveal that targeting ITGA5 using inhibitors that are currently under consideration in clinical trials may be beneficial for patients with hypoxic tumors. Mol Cancer Res; 15(6); 723-34. ©2017 AACR.


Assuntos
Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Integrina alfa5beta1/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias da Mama/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Integrina alfa5beta1/metabolismo , Integrina beta1/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Regiões Promotoras Genéticas , Esferoides Celulares/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cancer Res ; 76(22): 6458-6462, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27811150

RESUMO

Phosphoglycerate dehydrogenase (PHGDH) is the metabolic enzyme responsible for shunting the glycolytic intermediate 3-phosphoglycerate to the serine synthesis pathway. In breast cancer and several other types of cancer, increased PHGDH expression is associated with patient mortality. Early studies focused on the role of PHGDH in promoting cell proliferation in the small percentage of breast cancers with PHGDH gene amplification. However, recent studies have revealed a critical role for PHGDH and downstream enzymes of the serine synthesis pathway and one carbon metabolism in NADPH production and the maintenance of redox homeostasis, which are required for enrichment of breast cancer stem cells in response to hypoxia or chemotherapy. These results provide a mechanism for PHGDH overexpression in breast cancers in which PHGDH is not amplified and have implications for improving the response of triple-negative breast cancers to cytotoxic chemotherapy. Cancer Res; 76(22); 6458-62. ©2016 AACR.


Assuntos
Serina/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Humanos , Células-Tronco Neoplásicas/metabolismo , Oxirredução
16.
Cell Signal ; 28(10): 1537-44, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27424491

RESUMO

Claudin-4 has been identified as an integral member of tight junctions and has been found to be upregulated in various types of cancers especially in metastatic cancers. However, the molecular mechanism of the upregulation of Claudin-4 and its role in lung tumorigenesis are unknown. The aim of the present study is to investigate the role of Claudin-4 on migration and tumorigenicity of lung cancer cells and to examine the regulatory effects of TGF-ß on Claudin-4 expression. We have observed that TGF-ß induces the expression of Claudin-4 dramatically in lung cell lines in a time dependent manner. TGF-ß-induced Smad signaling is important for enhancing Claudin-4 mRNA level through inducing its promoter activity. Treatment with curcumin, a c-Jun inhibitor, or stable knockdown of c-Jun abrogates TGF-ß-induced Claudin-4 expression suggesting an involvement of the c-Jun pathway. Notably, TGF-ß-induced Claudin-4 expression through c-Jun pathway plays a role in TGF-ß-mediated motility and tumorigenicity of these cells. In support of these observations, we have uncovered that Claudin-4 is upregulated in 14 of 24 (58%) lung tumors when compared with normal lung tissue. This is the first study to show how TGF-ß regulates the expression of Claudin-4 through c-Jun signaling and how this pathway contributes to the migratory and tumorigenic phenotype of lung tumor cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Claudina-4/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Claudina-4/genética , Humanos , Neoplasias Pulmonares/patologia , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos
17.
Oncotarget ; 7(40): 64527-64542, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27590511

RESUMO

Exposure of breast cancer cells to hypoxia increases the percentage of breast cancer stem cells (BCSCs), which are required for tumor initiation and metastasis, and this response is dependent on the activity of hypoxia-inducible factors (HIFs). We previously reported that exposure of breast cancer cells to hypoxia induces the ALKBH5-mediated demethylation of N6-methyladenosine (m6A) in NANOG mRNA leading to increased expression of NANOG, which is a pluripotency factor that promotes BCSC specification. Here we report that exposure of breast cancer cells to hypoxia also induces ZNF217-dependent inhibition of m6A methylation of mRNAs encoding NANOG and KLF4, which is another pluripotency factor that mediates BCSC specification. Although hypoxia induced the BCSC phenotype in all breast-cancer cell lines analyzed, it did so through variable induction of pluripotency factors and ALKBH5 or ZNF217. However, in every breast cancer line, the hypoxic induction of pluripotency factor and ALKBH5 or ZNF217 expression was HIF-dependent. Immunohistochemistry revealed that expression of HIF-1α and ALKBH5 was concordant in all human breast cancer biopsies analyzed. ALKBH5 knockdown in MDA-MB-231 breast cancer cells significantly decreased metastasis from breast to lungs in immunodeficient mice. Thus, HIFs stimulate pluripotency factor expression and BCSC specification by negative regulation of RNA methylation.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Neoplasias da Mama/metabolismo , Células-Tronco Neoplásicas/fisiologia , RNA Mensageiro/genética , Transativadores/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Hipóxia Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Células MCF-7 , Metilação , Camundongos , Camundongos SCID , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Res ; 76(15): 4430-42, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27280394

RESUMO

Intratumoral hypoxia stimulates enrichment of breast cancer stem cells (BCSC), which are critical for metastasis and patient mortality. Here we report a metabolic adaptation that is required for hypoxia-induced BCSC enrichment and metastasis. Hypoxia-inducible factors coordinately regulate expression of genes encoding phosphoglycerate dehydrogenase (PHGDH) and five downstream enzymes in the serine synthesis pathway and mitochondrial one-carbon (folate) cycle. RNAi-mediated silencing of PHGDH expression in both estrogen receptor-positive and negative breast cancer cells led to decreased NADPH levels, disturbed mitochondrial redox homeostasis, and increased apoptosis, which abrogated BCSC enrichment under hypoxic conditions. PHGDH-deficient cells exhibited increased oxidant levels and apoptosis, as well as loss of BCSC enrichment, in response to treatment with carboplatin or doxorubicin. PHGDH-deficient cells were relatively weakly tumorigenic and tumors that did form were deficient in BCSCs, abolishing metastatic capacity. Our findings highlight a role for PHGDH in the formation of secondary (recurrent or metastatic) tumors, with potential implications for therapeutic targeting of advanced cancers. Cancer Res; 76(15); 4430-42. ©2016 AACR.


Assuntos
Neoplasias da Mama/genética , Neoplasias Pulmonares/secundário , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fosfoglicerato Desidrogenase/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Transfecção
19.
Oncotarget ; 5(24): 12509-27, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25587023

RESUMO

Intratumoral hypoxia, which is associated with breast cancer metastasis and patient mortality, increases the percentage of breast cancer stem cells (BCSCs) but the underlying molecular mechanisms have not been delineated. Here we report that hypoxia-inducible factor 1 (HIF-1) triggers the expression and activity of TAZ, a transcriptional co-activator that is required for BCSC maintenance, through two discrete mechanisms. First, HIF-1 binds directly to the WWTR1 gene and activates transcription of TAZ mRNA. Second, HIF-1 activates transcription of the SIAH1 gene, which encodes a ubiquitin protein ligase that is required for the hypoxia-induced ubiquitination and proteasome-dependent degradation of LATS2, a kinase that inhibits the nuclear localization of TAZ. Inhibition of HIF-1α, TAZ, or SIAH1 expression by short hairpin RNA blocked the enrichment of BCSCs in response to hypoxia. Human breast cancer database analysis revealed that increased expression (greater than the median) of both TAZ and HIF-1 target genes, but neither one alone, is associated with significantly increased patient mortality. Taken together, these results establish a molecular mechanism for induction of the BCSC phenotype in response to hypoxia.


Assuntos
Neoplasias da Mama/patologia , Fator 1 Induzível por Hipóxia/metabolismo , Fatores de Transcrição/biossíntese , Aciltransferases , Animais , Núcleo Celular/metabolismo , Feminino , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Camundongos SCID , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
Front Biosci (Landmark Ed) ; 17(4): 1281-93, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22201803

RESUMO

Members of the TGF-beta superfamily exhibit various biological activities, and perturbations of their signaling are linked to certain clinical disorders including cancer. The role of TGF-beta signaling as a tumor suppressor pathway is best illustrated by the presence of inactivating mutations in genes encoding TGF-beta receptors and Smads in human carcinomas. This perspective is further supported by studies of tumor development in mouse models after modulation of receptors and Smads. TGF-beta also controls processes such as cell invasion, immune regulation, and microenvironment alterations that cancer cells may exploit to their advantage for their progression. Consequently, the output of a TGF-beta response is highly situation dependent, across different tissues, and also in cancer in general. Understanding the mechanisms of TGF-beta superfamily signaling is thus important for the development of new ways to treat various types of cancer. This review focuses on recent advances in understanding the Smad dependent TGF-beta pathway as it relates to human carcinogenesis.


Assuntos
Neoplasias/metabolismo , Proteínas Smad/metabolismo , Animais , Humanos , Camundongos , Modelos Moleculares , Fator de Crescimento Transformador beta/antagonistas & inibidores , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA