Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 8(62): 104894-104912, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29285221

RESUMO

Triple negative breast cancers (TNBC) have poor prognosis compared to other breast cancer subtypes and represent 15-20% of breast cancers diagnosed. Unique targets and new molecularly-targeted therapies are urgently needed for this subtype. Despite high expression of Epidermal Growth Factor Receptor, inhibitors such as lapatinib have not shown therapeutic efficacy in TNBC patients. Herein, we report that treatment with the covalent JNK inhibitor, JNK-IN-8, synergizes with lapatinib to cause cell death, while these compounds as single agents have little effect. The combination significantly increases survival of mice bearing xenografts of MDA-MB-231 human TNBC cells. Our studies demonstrate that lapatinib treatment increases c-Jun and JNK phosphorylation indicating a mechanism of resistance. Combined, these compounds significantly reduce transcriptional activity of Nuclear Factor kappa B, Activating Protein 1, and Nuclear factor erythroid 2-Related Factor 2. As master regulators of antioxidant response, their decreased activity induces a 10-fold increase in reactive oxygen species that is cytotoxic, and is rescued by addition of exogenous antioxidants. Over expression of p65 or Nrf2 also significantly rescues viability during JNK-IN-8 and lapatinib treatment. Further studies combining JNK-IN-8 and lapatinib may reveal a benefit for patients with TNBC, fulfilling a critical medical need.

2.
ACS Med Chem Lett ; 8(10): 1072-1076, 2017 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-29057053

RESUMO

Serotonin (5-hydroxytryptamine, 5-HT) is a critical local regulator of epithelial homeostasis in the breast and exerts its actions through a number of receptors. Dysregulation of serotonin signaling is reported to contribute to breast cancer pathophysiology by enhancing cell proliferation and promoting resistance to apoptosis. Preliminary analyses indicated that the potent 5-HT1B/1D serotonin receptor agonist 5-nonyloxytryptamine (5-NT), a triptan-like molecule, induced cell death in breast cancer cell lines. Thus, we synthesized a series of novel alkyloxytryptamine analogues, several of which decreased the viability of various human cancer cell lines. Proteomic and metabolomic analyses showed that compounds 6 and 10 induced apoptosis and interfered with signaling pathways that regulate protein translation and survival, such as the Akt/mTOR pathway, in triple-negative breast cancer cells.

3.
Elife ; 42015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-26030852

RESUMO

Ataxia-telangiectasia mutated (ATM) protein kinase regulates the DNA damage response (DDR) and is associated with cancer suppression. Here we report a cancer-promoting role for ATM. ATM depletion in metastatic cancer cells reduced cell migration and invasion. Transcription analyses identified a gene network, including the chemokine IL-8, regulated by ATM. IL-8 expression required ATM and was regulated by oxidative stress. IL-8 was validated as an ATM target by its ability to rescue cell migration and invasion defects in ATM-depleted cells. Finally, ATM-depletion in human breast cancer cells reduced lung tumors in a mouse xenograft model and clinical data validated IL-8 in lung metastasis. These findings provide insights into how ATM activation by oxidative stress regulates IL-8 to sustain cell migration and invasion in cancer cells to promote metastatic potential. Thus, in addition to well-established roles in tumor suppression, these findings identify a role for ATM in tumor progression.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Neoplasias da Mama/metabolismo , Movimento Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Interleucina-8/metabolismo , Neoplasias Pulmonares/metabolismo , Invasividade Neoplásica/fisiopatologia , Estresse Oxidativo/fisiologia , Animais , Western Blotting , Fracionamento Celular , Imunoprecipitação da Cromatina , Primers do DNA/genética , Eletroforese em Gel de Poliacrilamida , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/fisiologia , Humanos , Luciferases , Neoplasias Pulmonares/secundário , Camundongos , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real
4.
Genes Cancer ; 4(9-10): 378-87, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24349635

RESUMO

Disseminated cancer cells rely on intricate interactions among diverse cell types in the tumor-associated stroma, vasculature, and immune system for survival and growth. Ubiquitous expression of c-Jun N-terminal kinase (jnk) genes in various cell types permits their control of metastasis. In early stages of metastasis, JNKs affect tumor-associated inflammation and angiogenesis as well as tumor cell migration and intravasation. Within the tumor stroma, JNKs are essential for the release of growth factors that promote epithelial-to-mesenchymal transition (EMT) in tumor cells. JNK3, the least ubiquitous isoform, facilitates angiogenesis by increasing endothelial cell migration. Importantly, JNK expression in tumor cells integrates stromal signals to promote tumor cell invasion. However, JNK isoforms differentially regulate migration toward the endothelial barrier. Once tumor cells enter the bloodstream, JNKs increase circulating tumor cell (CTC) survival and homing to tissues. By promoting fibrosis, JNKs improve CTC attachment to the endothelium. Once anchored, JNKs stimulate EMT to facilitate tumor cell extravasation and enhance the secretion of endothelial barrier disrupters. Tumor cells attract barrier-disrupting macrophages by JNK-dependent transcription of macrophage chemoattractant molecules. In the secondary tissue, JNKs are instrumental in the premetastatic niche and stimulate tumor cell proliferation. JNK expression in cancer cells stimulates tissue-remodeling macrophages to improve tumor colonization. However, in T-cells, JNKs alter cytokine production that increases tumor surveillance and inhibits the recruitment of tissue-remodeling macrophages. Therapeutically targeting JNKs for metastatic disease is attractive considering their promotion of metastasis; however, specific JNK tools are needed to determine their definitive actions within the context of the entire metastatic cascade.

5.
ACS Chem Biol ; 6(6): 658-66, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21438496

RESUMO

Despite their lack of selectivity toward c-Jun N-terminal kinase (JNK) isoforms, peptides derived from the JIP (JNK Interacting Protein) scaffolds linked to the cell-penetrating peptide TAT are widely used to investigate JNK-mediated signaling events. To engineer an isoform-selective peptide inhibitor, several JIP-based peptide sequences were designed and tested. A JIP sequence connected through a flexible linker to either the N-terminus of an inverted TAT sequence (JIP(10)-Δ-TAT(i)) or to a poly arginine sequence (JIP(10)-Δ-R(9)) enabled the potent inhibition of JNK2 (IC(50) ≈ 90 nM) and exhibited 10-fold selectivity for JNK2 over JNK1 and JNK3. Examination of both peptides in HEK293 cells revealed a potent ability to inhibit the induction of both JNK activation and c-Jun phosphorylation in cells treated with anisomycin. Notably, Western blot analysis indicates that only a fraction of total JNK must be activated to elicit robust c-Jun phosphorylation. To examine the potential of each peptide to selectively modulate JNK2 signaling in vivo, their ability to inhibit the migration of Polyoma Middle-T Antigen Mammary Tumor (PyVMT) cells was assessed. PyVMTjnk2-/- cells exhibit a lower migration potential compared to PyVMTjnk2+/+ cells, and this migration potential is restored through the overexpression of GFP-JNK2α. Both JIP(10)-Δ-TAT(i) and JIP(10)-Δ-R(9) inhibit the migration of PyVMTjnk2+/+ cells and PyVMTjnk2-/- cells expressing GFP-JNK2α. However, neither peptide inhibits the migration of PyVMTjnk2-/- cells. A control form of JIP(10)-Δ-TAT(i) containing a single leucine to arginine mutation lacks ability to inhibit JNK2 in vitro cell-free and cell-based assays and does not inhibit the migration of PyVMTjnk2+/+ cells. Together, these data suggest that JIP(10)-Δ-TAT(i) and JIP(10)-Δ-R(9) inhibit the migration of PyVMT cells through the selective inhibition of JNK2. Finally, the mechanism of inhibition of a D-retro-inverso JIP peptide, previously reported to inhibit JNK, was examined and found to inhibit p38MAPKα in an in vitro cell-free assay with little propensity to inhibit JNK isoforms.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Peptídeos/farmacologia , Animais , Neoplasias da Mama/enzimologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Peptídeos/química , Peptídeos/isolamento & purificação , Relação Estrutura-Atividade , Especificidade por Substrato , Células Tumorais Cultivadas
6.
PLoS One ; 5(5): e10443, 2010 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-20454618

RESUMO

Oncogenes induce cell proliferation leading to replicative stress, DNA damage and genomic instability. A wide variety of cellular stresses activate c-Jun N-terminal kinase (JNK) proteins, but few studies have directly addressed the roles of JNK isoforms in tumor development. Herein, we show that jnk2 knockout mice expressing the Polyoma Middle T Antigen transgene developed mammary tumors earlier and experienced higher tumor multiplicity compared to jnk2 wildtype mice. Lack of jnk2 expression was associated with higher tumor aneuploidy and reduced DNA damage response, as marked by fewer pH2AX and 53BP1 nuclear foci. Comparative genomic hybridization further confirmed increased genomic instability in PyV MT/jnk2-/- tumors. In vitro, PyV MT/jnk2-/- cells underwent replicative stress and cell death as evidenced by lower BrdU incorporation, and sustained chromatin licensing and DNA replication factor 1 (CDT1) and p21(Waf1) protein expression, and phosphorylation of Chk1 after serum stimulation, but this response was not associated with phosphorylation of p53 Ser15. Adenoviral overexpression of CDT1 led to similar differences between jnk2 wildtype and knockout cells. In normal mammary cells undergoing UV induced single stranded DNA breaks, JNK2 localized to RPA (Replication Protein A) coated strands indicating that JNK2 responds early to single stranded DNA damage and is critical for subsequent recruitment of DNA repair proteins. Together, these data support that JNK2 prevents replicative stress by coordinating cell cycle progression and DNA damage repair mechanisms.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Replicação do DNA , Instabilidade Genômica , Neoplasias Mamárias Animais/enzimologia , Neoplasias Mamárias Animais/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Estresse Fisiológico , Aneuploidia , Animais , Cafeína/farmacologia , Morte Celular/efeitos dos fármacos , Proteínas Cromossômicas não Histona , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Feminino , Fase G1/efeitos dos fármacos , Amplificação de Genes/efeitos dos fármacos , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Lesões Pré-Cancerosas/enzimologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Proteína de Replicação A/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Transgenes/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
7.
J Biol Chem ; 283(42): 28265-73, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18697743

RESUMO

Significant discoveries have recently contributed to our knowledge of intracellular growth factor and nutrient signaling via mTOR (mammalian target of rapamycin). This signaling pathway is essential in cellular metabolism and cell survival by enhancing protein translation through phosphorylation of 4EBP-1 and p70S6K. Growth factors like insulin-like growth factor-I induce mTOR to prevent cell death during cellular stress. Agents targeting mTOR are of major interest as anticancer agents. We show here, using human breast cancer cells, that certain types of stress activate mTOR leading to 4E-BP1 and p70S6K phosphorylation. UV treatment increased phosphorylation of the translation inhibitor eIF2alpha, suggesting a potential mechanism for UV activation of Akt and mTOR. c-Myc, a survival protein regulated by cap-dependent protein translation, increased with IGF-I treatment, but this response was not inhibited by rapamycin. Additionally, UV treatment potently increased c-Myc degradation, which was reduced by co-treatment with the proteasomal inhibitor, MG-132. Together, these data suggest that protein translation does not strongly mediate cell survival in these models. In contrast, the phosphorylation status of retinoblastoma protein (pRB) was mediated by mTOR through its inhibitory effects on phosphatase activity. This effect was most notable during DNA damage and rapamycin treatment. Hypophosphorylated pRB was susceptible to inactivation by caspase-mediated cleavage, resulting in cell death. Reduction of pRB expression inhibited IGF-I survival effects. Our data support an important role of phosphatases and pRB in IGF-I/mTOR-mediated cell survival. These studies provide new directions in optimizing anticancer efficacy of mTOR inhibitors when used in combination with DNA-damaging agents.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Inibidores Enzimáticos/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Leupeptinas/farmacologia , Modelos Biológicos , Fosforilação , Inibidores de Proteassoma , Proteínas Proto-Oncogênicas c-myc/metabolismo , Estresse Fisiológico , Serina-Treonina Quinases TOR , Raios Ultravioleta
8.
Breast Cancer Res Treat ; 91(3): 259-69, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15952059

RESUMO

IRS-1 (Insulin Receptor Substrate-1) is an adaptor protein important for insulin and IGF-I receptor (Insulin-like Growth Factor-IR) transduction to downstream targets. One mechanism recently identified to downregulate IGF-I or insulin receptor signaling in diabetic models is IRS-1 Ser(312) phosphorylation. To date, the importance of this residue in cancer is unknown. This paper identifies mechanisms leading to Ser(312) regulation in MCF-7 breast cancer cells. Whereas IGF-I phosphorylation of IRS(312) is PI (phosphatidylinositol) 3-kinase dependent, anisomycin stress treatment requires JNK activation to induce phosphorylation of IRS(312). We show that both IGF-I and anisomycin stress treatment converge downstream onto mTOR (Mammalian Target of Rapamycin) and PKCdelta (Protein Kinase C-delta) to induce IRS-1 Ser(312) phosphorylation. mTOR associates with IRS-1 and is primarily required for Ser(312) phosphorylation in response to stress or IGF-I treatment. PKCdelta binds to mTOR and its activity is also important for stress or IGF-I mediated Ser(312) phosphorylation. Thus, mTOR and PKCdelta convey diverse signals to regulate IRS-1 function.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Anisomicina/farmacologia , Neoplasias da Mama/patologia , Ativação Enzimática , Feminino , Humanos , Proteínas Substratos do Receptor de Insulina , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Proteína Quinase C-delta , Proteínas Quinases/química , Proteínas Quinases/genética , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina/química , Transdução de Sinais , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas , Tirosina/metabolismo
9.
Oncogene ; 23(2): 596-604, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14724588

RESUMO

c-Jun N-terminal kinase (JNK) is activated by diverse cell stimuli, including stress, growth factors, and cytokines. Traditionally, activation of JNK by stress treatment is thought to induce cell death. However, our recent data indicate that JNK's ability to sensitize cells to apoptosis may be, in part, cell cycle dependent. Here, we show that the majority of both paclitaxel- and UV-induced apoptosis can be inhibited by the pharmacological JNK inhibitor, SP600125, in MCF-7 cells. However, inhibition of JNK does little to reverse doxorubicin-induced apoptosis in MCF-7 cells or doxorubicin- and UV-mediated death in MDA MB-231 cells. SP treatment causes G2/M arrest of three breast cancer cell lines and results in the endoreduplication (cellular DNA content >4N) of MCF-7 and MDA MB-231 cells. These effects on cell cycle and apoptosis are not significantly altered by the inhibition of p53, indicating that JNK is functioning independently of p53. Lastly, inhibition of JNK using both SP and antisense oligonucleotides targeted to JNK1 and JNK2 reduced proliferation of all three breast cancer cell lines. Taken together, these results suggest that the activation of JNK is important for the induction of apoptosis following stresses that function at different cell cycle phases, and that basal JNK activity is necessary to promote proliferation and maintain diploidy in breast cancer cells.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Replicação do DNA , Fase G2 , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mitose , Proteína Supressora de Tumor p53/metabolismo , Antracenos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Neoplasias da Mama/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Linhagem Celular Tumoral , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Doxorrubicina/farmacologia , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose/efeitos dos fármacos , Mitose/efeitos da radiação , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Paclitaxel/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Raios Ultravioleta
10.
J Biol Chem ; 278(46): 45793-800, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12954616

RESUMO

The importance of the mitochondria in UV-induced apoptosis has become increasingly apparent. Following DNA damage cytochrome c and other pro-apoptotic factors are released from the mitochondria, allowing for formation of the apoptosome and subsequent cleavage and activation of caspase-9. Active caspase-9 then activates downstream caspases-3 and/or -7, which in turn cleave poly(ADP)-ribose polymerase (PARP) and other down-stream targets, resulting in apoptosis. In an effort to understand the mechanisms of Akt-mediated cell survival in breast cancer, we studied the effects of insulin-like growth factor (IGF)-I treatment on UV-treated MCF-7 human breast cancer cells. Apoptosis was induced in MCF-7 cells after UV treatment, as measured by caspase-7 and PARP cleavage, and IGF-I co-treatment protected against this response. Surprisingly caspase-9 cleavage was unchanged with UV and/or IGF-I treatment. Using MCF-7 cells overexpressing caspase-3 we have shown that resistance of caspase-9 to cleavage was not altered by the expression of caspase-3. Furthermore, overexpression of caspase-9 did not enhance PARP or caspase-7 cleavage after UV treatment. Because caspase-8 was activated with UV treatment alone, we believe that UV-induced apoptosis in MCF-7 cells occurs independently of cytochrome c and caspase-9, supporting the existence of a cytoplasmic inhibitor of cytochrome c in MCF-7 cells. We anticipate that such inhibitors may be overexpressed in cancer cells, allowing for treatment resistance.


Assuntos
Apoptose , Caspases/fisiologia , Citocromos c/metabolismo , Western Blotting , Neoplasias da Mama/metabolismo , Caspase 3 , Caspase 7 , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Microscopia de Fluorescência , Fosfatidilinositol 3-Quinases/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Testes de Precipitina , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA/metabolismo , Fatores de Tempo , Transfecção , Raios Ultravioleta , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
11.
Oncogene ; 22(4): 602-14, 2003 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-12555073

RESUMO

Insulin-like growth factor-I receptor (IGF-IR) is frequently overexpressed in a variety of cancer types. Since many breast tumors and cancer cell lines overexpress IGF-IR, we tested IGF-I effects on chemotherapy-treated breast cancer cells. IGF-I protects from chemotherapy-induced apoptosis, suggesting that overlapping signaling pathways modulate IGF-I and chemotherapy treatment outcomes. Taxol and other chemotherapy drugs induce c-Jun N-terminal kinase (JNK), a kinase that conveys cellular stress and death signals. Notably, in this paper we show that IGF-I alone induces a potent JNK response and this activity is reversed by inhibition of phosphatidylinositol 3-kinase (PI 3-kinase) with LY294002 in MCF-7 but not T47D cells. Cotreatment of cells with chemotherapy and IGF-I leads to additive JNK responses. Using cells overexpressing Akt, we confirm that IGF-I-mediated survival is Akt dependent. In contrast, overexpression of JNK significantly enhances Taxol-induced apoptosis and inhibits IGF-I survival effects. Further, JNK attenuates anchorage-independent growth of MCF-7 cells. The inhibitory effect of JNK appears to be mediated by serine phosphorylation of IRS-1 (insulin receptor substrate) since both Taxol and IGF-I treatment enhanced Ser(312) IRS-1 phosphorylation, while LY294002 blocked IGF-I-mediated phosphorylation. Taken together, these data provide a mechanism whereby stress or growth factors activate JNK to reduce proliferation and/or survival in breast cancer cells.


Assuntos
Neoplasias da Mama/enzimologia , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Transdução de Sinais , Sequência de Bases , Western Blotting , Neoplasias da Mama/patologia , Divisão Celular , Sobrevivência Celular , Primers do DNA , Humanos , Proteínas Substratos do Receptor de Insulina , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilação , Testes de Precipitina , Serina/metabolismo , Células Tumorais Cultivadas
12.
Breast Dis ; 17: 49-59, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15687677

RESUMO

The insulin-like growth factor system has been implicated in the proliferative control of breast cancer cells. In addition to this function, IGF action can also protect cells from programmed cell death. Substantial knowledge has been gained about death effector molecules and their regulation in breast cancer. IGF receptor can influence several key cell death pathways. In this review, we will focus on the intracellular mechanisms activated by IGF-I that influence cell survival.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...