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
1.
Cancers (Basel) ; 14(20)2022 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-36291767

RESUMO

TIMP-1 is one of the many factors that CAFs have been shown to secret. TIMP-1 can act in a tumor-supportive or tumor-suppressive manner. The purpose of this study was to elucidate the role of CAF-secreted TIMP-1 for the effects of CAFs on breast cancer cell behavior. Breast cancer cells were exposed to conditioned medium collected from TIMP-1-secreting CAFs (CAF-CM), and the specific effects of TIMP-1 on protein expression, migration and growth were examined using TIMP-1-specifc siRNA (siTIMP1), recombinant TIMP-1 protein (rhTIMP-1) and TIMP-1 level-rising phorbol ester. We observed that TIMP-1 increased the expression of its binding partner CD63 and induced STAT3 and ERK1/2 activation by cooperating with CD63 and integrin ß1. Since TIMP-1 expression was found to be dependent on STAT3, TIMP-1 activated its own expression, resulting in a TIMP-1/CD63/integrin ß1/STAT3 feedback loop. IL-6, a classical STAT3 activator, further fueled this loop. Knock-down of each component of the feedback loop prevented the CAF-induced increase in migratory activity and inhibited cellular growth in adherent cultures in the presence and absence of the anti-estrogen fulvestrant. These data show that TIMP-1/CD63/integrin ß1/STAT3 plays a role in the effects of CAFs on breast cancer cell behavior.

2.
Virchows Arch ; 480(3): 529-541, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35020071

RESUMO

Patients with estrogen receptor positive breast cancer are usually receiving an anti-estrogen therapy by either aromatase inhibitors or selective estrogen receptor mediators such as tamoxifen. Nevertheless, acquired resistance to tamoxifen under treatment frequently hampers therapy. One proposed explanation for this phenomenon is the interaction of the tumor cells with cells of the tumor microenvironment via the Insulin-like growth factor RNA binding protein 5/B-cell lymphoma 3 (IGFBP5/BCL3) axis. Here we investigated whether a high expression of BCL3 either cytoplasmic or nuclear is associated with the occurrence of a relapse under anti-estrogen therapy in patients. Formaldehyde-fixed, paraffin-embedded samples of 180 breast cancer patients were analyzed for BCL3 expression by immunohistochemistry. An immunoreactive score (IRS) was calculated from staining intensity in cytoplasm and nucleus as well as the percentage of positive tumor cells. These scores were correlated with clinico-pathological parameters using cross-tabulation analysis and patients' relapse free and overall survival by Kaplan-Meier analysis and Cox regression. A tamoxifen-adapted MCF-7 derived cell line was investigated for BCL3 localization by immunofluorescence. The cytosolic BCL3-IRS significantly correlated with the proliferation marker Ki-67, and with the occurrence of a relapse under tamoxifen treatment. Nuclear score correlated only with tamoxifen-relapse. In survival analysis, both scores were highly significant prognostic factors for relapse free, but not for overall survival. This was especially obvious for estrogen receptor positive and HER2/NEU negative cases as well as lobular breast cancer. Tamoxifen-treated, but not aromatase-treated patients had a poor survival when BCL3 scores were high. A tamoxifen adapted cell line exhibited a reduced expression and mainly nuclear localization of BCL3, compared to the parental estrogen receptor positive cell-line MCF-7. Altogether, these data strongly support a function of BCL3 in tamoxifen resistance and its potential use as a predictive biomarker for tamoxifen resistance.


Assuntos
Neoplasias da Mama , Tamoxifeno , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Recidiva Local de Neoplasia , Receptores de Estrogênio/metabolismo , Estudos Retrospectivos , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Microambiente Tumoral
3.
Cancers (Basel) ; 12(11)2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33228022

RESUMO

CAFs (Carcinoma-associated fibroblasts) play an important role in cancer progression. For instance, they promote resistance to anti-estrogens, such as fulvestrant. Here, we show that, in ERα-positive breast cancer cell lines, the cocktail of factors secreted by CAFs (CAF-CM) induce the expression of the embryonal stem cell transcription factor Sox2 (sex determining region Y (SRY)-box 2). Long-term exposure to CAF-CM was able to give rise to very high Sox2 levels both in the absence and presence of fulvestrant. IL-6 (interleukin-6), a major component of CAF-CM, failed to raise Sox2 expression. In MCF-7 sublines established in the presence of CAF-CM, almost all cells showed Sox2 expression, whereas long-term treatment of T47D cells with CAF-CM resulted in a ~60-fold increase in the proportions of two distinct populations of Sox2 high and low expresser cells. Exposure of BT474 cells to CAF-CM raised the fraction of Sox2 high expresser cells by ~3-fold. Cell sorting based on CD44 and CD24 expression or ALDH (aldehyde dehydrogenase) activity revealed that most Sox2 high expresser cells were not CD44hi/CD24lo- or ALDH-positive cells suggesting that they were not CSCs (cancer stem cells), though CD44 played a role in Sox2 expression. Functionally, Sox2 was found to protect CAF-CM-treated cells against apoptosis and to allow higher growth activity in the presence of fulvestrant. Mechanistically, the key drivers of Sox2 expression was found to be STAT3 (Signal transducer and activator of transcription 3), Bcl-3 (B-cell lymphoma 3) and the PI3K (Phosphoinositide 3-kinase)/AKT pathway, whose activities/expression can all be upregulated by CAF-CM. These data suggest that CAF-CM induces Sox2 expression in non-CSCs by activating proteins involved in growth control and drug resistance, leading to higher protection against apoptosis.

4.
Int J Oncol ; 56(1): 258-272, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31789400

RESUMO

Carcinoma­associated fibroblasts (CAFs) secrete factors that increase the expression and/or activities of proteins in breast cancer cells and induce resistance to anti­estrogens, such as fulvestrant. A major factor is interleukin­6 (IL­6). This study demonstrated that, across estrogen receptor (ER)α­positive and ­negative cell lines, recombinant human IL­6 (rhIL­6) mimicked most of the CAF­conditioned medium (CM)­induced changes in protein expression patterns; however, in most cases, it failed to recapitulate CAF­CM­triggered alterations in ERK1/2 and AKT activities. The ability of rhIL­6 to induce fulvestrant resistance was dependent upon the culture conditions. In 3D, but not in 2D cultures, rhIL­6 increased the survival of fulvestrant­treated cells, although not to the same extent as observed with CAF­CM. In 2D cultures, rhIL­6 acted in a pro­apoptotic manner and decreased the expression of ATP­binding cassette transporter G2 (ABCG2). The inhibition of the PI3K/AKT pathway had similar effects on apoptosis and ABCG2 expression, linking the failure of rhIL­6 to induce fulvestrant resistance to its inability to activate the PI3K/AKT pathway. In 3D cultures, both CAF­CM and rhIL­6 acted in an anti­apoptotic manner. These activities are likely independent on the PI3K/AKT pathway and ABCG2. Experiments on ERα­negative breast cancer cells revealed a growth­inhibitory effects of both CAF­CM and rhIL­6, which coincided with a reduction in the c­Myc level. These data suggest that IL­6 plays a role in several effects of CAF­CM, including alterations in protein expression patterns, fulvestrant resistance in 3D cultures and growth inhibition. By contrast, IL­6 is unlikely to be responsible for the CAF­CM­induced activation of the PI3K/AKT pathway and fulvestrant resistance in 2D cultures.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/patologia , Proliferação de Células , Meios de Cultivo Condicionados/farmacologia , Resistencia a Medicamentos Antineoplásicos , Fulvestranto/farmacologia , Interleucina-6/metabolismo , Antineoplásicos Hormonais/farmacologia , Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Feminino , Humanos , Interleucina-6/genética , Células Tumorais Cultivadas
5.
Oncotarget ; 9(31): 22079-22094, 2018 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-29774124

RESUMO

We studied the long-term effect of stromal factors on the development of fulvestrant-resistance (FR) and fulvestrant-induced dormancy (D). Sublines established from stroma-treated FR-cells (C-FR cells) and D-cells (C-D cells) show permanently high expression of integrin ß1 as well as Bcl-3 and P-STAT3 (C-FR) or IGF1R (C-D). Yet, cells fail to withstand fulvestrant better and do not migrate or grow faster than control cells. Instead, C-D cells rely on stromal factors to perform as well as control cells. In addition, C-FR cells adapted to integrin ß1 for growth in 3D cultures. These data suggest that long-term exposure to stromal factors leads to addiction rather than better performance in cellular activities. We also found that morphologically distinct breast cancer cell line subpopulations share key responses to stromal factors suggesting that intratumoral heterogeneity may play a minor role in the interaction between breast cancer and stromal cells.

6.
Oncotarget ; 7(25): 38408-38426, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27224909

RESUMO

We have previously shown that stromal cells desensitize breast cancer cells to the anti-estrogen fulvestrant and, along with it, downregulate the expression of TMEM26 (transmembrane protein 26). In an effort to study the function and regulation of TMEM26 in breast cancer cells, we found that breast cancer cells express non-glycosylated and N-glycosylated isoforms of the TMEM26 protein and demonstrate that N-glycosylation is important for its retention at the plasma membrane. Fulvestrant induced significant changes in expression and in the N-glycosylation status of TMEM26. In primary breast cancer, TMEM26 protein expression was higher in ERα (estrogen receptor α)/PR (progesterone receptor)-positive cancers. These data suggest that ERα is a major regulator of TMEM26. Significant changes in TMEM26 expression and N-glycosylation were also found, when MCF-7 and T47D cells acquired fulvestrant resistance. Furthermore, patients who received aromatase inhibitor treatment tend to have a higher risk of recurrence when tumoral TMEM26 protein expression is low. In addition, TMEM26 negatively regulates the expression of integrin ß1, an important factor involved in endocrine resistance. Data obtained by spheroid formation assays confirmed that TMEM26 and integrin ß1 can have opposite effects in breast cancer cells. These data are consistent with the hypothesis that, in ERα-positive breast cancer, TMEM26 may function as a tumor suppressor by impeding the acquisition of endocrine resistance. In contrast, in ERα-negative breast cancer, particularly triple-negative cancer, high TMEM26 expression was found to be associated with a higher risk of recurrence. This implies that TMEM26 has different functions in ERα-positive and -negative breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proteínas de Membrana/biossíntese , Biomarcadores Farmacológicos/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/biossíntese , Feminino , Fulvestranto , Humanos , Integrina beta1/biossíntese , Células MCF-7 , Proteínas de Membrana/genética , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , RNA/genética , RNA/metabolismo
7.
Oncotarget ; 6(36): 39307-28, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26515727

RESUMO

There is strong evidence that stromal cells promote drug resistance of cancer. Here, we show that mesenchymal stem cells (MSCs) and carcinoma-associated fibroblasts (CAFs) desensitize ERα-positive breast cancer cells to the anti-estrogen fulvestrant. In search for the mechanism, we found that MSCs and CAFs similarly increased the activity of the PI3K/AKT and the JAK/STAT3 pathways and upregulated the expression of integrin ß1, IGF1R, HIF1α, CAIX and Bcl-3 in MCF-7 cells. Further analyses revealed that MSCs and CAFs coordinately induce these changes by triggering the downregulation of IGFBP5. Loss of IGFBP5 in MCF-7 cells was an early and long-lasting event in response to MSCs and CAFs and was accompanied by growth stimulation both in the absence and presence of fulvestrant. The growth-stimulatory effect in the absence of fulvestrant could be attributed to PI3K/AKT pathway activation and could be mimicked by insulin. The growth-promoting effect in the presence of fulvestrant depended upon the upregulation of Bcl-3. By cRNA microarray analysis we identified additional IGFBP5 targets, of which two (KLHL4 and SEPP1) were inversely regulated by IGFBP5 and Bcl-3. BT474 cells also responded to stromal cells by downregulating IGFBP5 and upregulating the P-AKT, Bcl-3 and IGF1R levels, whereas T47D cells did not show any of these responses. In conclusion, our data suggest that, by targeting IGFBP5 expression in ERα-positive breast cancer cells, such as MCF-7 cells, MSCs and CAFs are able to orchestrate a variety of events, particularly activation of the PI3K/AKT pathway, upregulation of Bcl-3 expression and desensitization to anti-estrogen.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Antagonistas do Receptor de Estrogênio/farmacologia , Fibroblastos/patologia , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Células-Tronco Mesenquimais/patologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Proteína 3 do Linfoma de Células B , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Resistencia a Medicamentos Antineoplásicos , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Humanos , Células MCF-7 , Transdução de Sinais
8.
PLoS One ; 8(1): e54261, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349840

RESUMO

Cellular functions are regulated by complex networks of many different signaling pathways. The TGFß and cAMP pathways are of particular importance in tumor progression. We analyzed the cross-talk between these pathways in breast cancer cells in 2D and 3D cultures. We found that cAMP potentiated TGFß-dependent gene expression by enhancing Smad3 phosphorylation. Higher levels of total Smad3, as observed in 3D-cultured cells, blocked this effect. Two Smad3 regulating proteins, YAP (Yes-associated protein) and TßRI (TGFß receptor 1), were responsive to cAMP. While YAP had little effect on TGFß-dependent expression and Smad3 phosphorylation, a constitutively active form of TßRI mimicked the cAMP effect on TGFß signaling. In 3D-cultured cells, which show much higher levels of TßRI and cAMP, TßRI was unresponsive to cAMP. Upregulation of TßRI expression by cAMP was dependent on transcription. A proximal TßRI promoter fragment was moderately, but significantly activated by cAMP suggesting that cAMP increases TßRI expression at least partially by activating TßRI transcription. Neither the cAMP-responsive element binding protein (CREB) nor the TßRI-regulating transcription factor Six1 was required for the cAMP effect. An inhibitor of histone deacetylases alone or together with cAMP increased TßRI expression by a similar extent as cAMP alone suggesting that cAMP may exert its effect by interfering with histone acetylation. Along with an additive stimulatory effect of cAMP and TGFß on p21 expression an additive inhibitory effect of these agents on proliferation was observed. Finally, we show that mesenchymal stem cells that interact with breast cancer cells can simultaneously activate the cAMP and TGFß pathways. In summary, these data suggest that combined effects of cAMP and TGFß, as e.g. induced by mesenchymal stem cells, involve the upregulation of TßRI expression on the transcriptional level, likely due to changes in histone acetylation. As a consequence, cancer cell functions such as proliferation are affected.


Assuntos
AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Colforsina/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fatores de Transcrição , Proteínas de Sinalização YAP
9.
Biochem Pharmacol ; 83(4): 480-8, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22142888

RESUMO

PI3K/Akt/mTOR and p53 signaling pathways are frequently deregulated in tumors. The anticancer drug RAD001 (everolimus) is a known mTOR-inhibitor, but mTOR-inhibition leads to phosphorylation of Akt inducing resistance against RAD001 treatment. There is growing evidence that conflicting signals transduced by the oncogene Akt and the tumorsuppressor p53 are integrated via negative feedback between the two pathways. We previously showed that the anti-malarial Chloroquine, a 4-alkylamino substituted quinoline, is a p53 activator and reduced the incidence of breast tumors in animal models. Additionally, Chloroquine is an effective chemosensitizer when used in combination with PI3K/Akt inhibitors but the mechanism is unknown. Therefore, our aim was to test, if Chloroquine could inhibit tumor growth and prevent RAD001-induced Akt activation. Chloroquine and RAD001 caused G1 cell cycle arrest in luminal MCF7 but not in mesenchymal MDA-MB-231 breast cancer cells, they significantly reduced MCF7 cell proliferation on a collagen matrix and mammospheroid formation. In a murine MCF7 xenograft model, combined treatment of Chloroquine and RAD001 significantly reduced mammary tumor growth by 4.6-fold (p = 0.0002) compared to controls. Chloroquine and RAD001 inhibited phosphorylation of mTOR and its downstream target, S6K1. Furthermore, Chloroquine was able to block the RAD001-induced phosphorylation of Akt serine 473. The Chloroquine effect of overcoming the RAD001-induced activation of the oncogene Akt, as well as the promising antitumor activity in our mammary tumor animal model present Chloroquine as an interesting combination partner for the mTOR-inhibitor RAD001.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cloroquina/farmacologia , Proteína Oncogênica v-akt/metabolismo , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Everolimo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Proteína Oncogênica v-akt/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/genética
10.
Oncol Rep ; 26(5): 1121-5, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21811762

RESUMO

Several members of the Ets (E26 transformation specific) transcription factor family are involved in tumor progression, e.g. by activating matrix metalloproteases. Ets proteins share a unique DNA-binding domain, the Ets domain, which specifically recognizes GGAA/T-containing sequences common in many promoters. While the roles of quite a number of Ets proteins in carcinogenesis have been well established, little is known about the importance of the Ets protein Elf-1 (E74-like factor 1) in cancer. Herein, we analyzed the expression of Elf-1 in breast cancer. We found that, like T-cells, breast cancer cells express both the 80 and 98 kDa isoforms of the Elf-1 protein with the 98 kDa isoform only be present in the nucleus. Immunohistochemical analysis of 119 breast cancer biopsies showed anti-Elf-1 immunoreactivity exclusively in the nucleus. Elf-1 expression varied largely among the breast cancer samples showing a negative correlation with histological grading. However, no association of Elf-1 expression with clinical outcome was observed, even when sub-cohorts of patients who received either only adjuvant endocrine treatment or only chemotherapy were separately analyzed. These data suggest that Elf-1 may modulate breast cancer progression to some extent without having an impact on survival of breast cancer patients.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Resultado do Tratamento
11.
Int J Oncol ; 39(3): 689-96, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21667024

RESUMO

Stromal cells, such as mesenchymal stem cells (MSCs) and carcinoma-associated fibroblasts (CAFs), play a role in cancer progression. To analyze their ability to modulate drug response, we generated spheroids of MCF-7 or MDA-MB-231 breast cancer cells in the absence or presence of human (h)MSCs or hCAFs and tested the susceptibility of the breast cancer cells to three different kinase inhibitors (TKI258, RAD001 and RAF265) used in cancer therapy. While stromal cells did not affect the response of either breast cancer cell line to the PDGFR/FGFR/VEGFR inhibitor TKI258, they sensitized breast cancer cells to the mTOR inhibitor RAD001. In MCF-7 cells, this was accompanied by increased apoptosis. hMSCs and to a lesser extent hCAFs also enhanced the cytotoxic effect of RAF inhibitor RAF265 on MDA-MB-231 cells. Searching for the mechanism that underlies the effect of stromal cells on RAF265 response we found that stromal cells inhibited RAF265-induced increase in ERK1/2 phosphorylation, supported RAF265-dependent downregulation of PKCα (protein kinase Cα) and prevented RAF265-induced conversion of LC3B, a marker of autophagy. To mimic the changes in ERK1/2 phosphorylation and PKCα expression in response to the stromal cells, we treated cells with MEK1 inhibitor U0126 or PKCα inhibitor Gö6976, respectively. U0126, but not Gö6976, was as effective as hMSCs in sensitizing MDA-MB-231 cells to RAF265. This suggests that hMSCs and hCAFs increased the cytotoxic effect of RAF265 on MDA-MB-231 cells by downregulating ERK1/2 phosphorylation. In summary, this study shows that hMSCs are able to render breast cancer cells more susceptible to kinase inhibitors and that, to the most part, hCAFs to which hMSCs can differentiate are able to mimic the drug-sensitizing effects of hMSCs.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Comunicação Celular/fisiologia , Fibroblastos/patologia , Células-Tronco Mesenquimais/patologia , Inibidores de Proteínas Quinases/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Everolimo , Feminino , Humanos , Imidazóis/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Células Estromais/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Quinases raf/antagonistas & inibidores
12.
Int J Oncol ; 36(2): 379-86, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20043072

RESUMO

Rho-GDIalpha is an inhibitor of Rho-GTPases, which is involved in cancer progression. Little is known about its role in breast cancer progression. There is evidence, that Rho-GDIalpha may modulate drug resistance of breast cancer cells. To assess the importance of Rho-GDIalpha as a risk factor in invasive ductal breast cancer, cancer specimens of three groups of patients were analyzed for Rho-GDIalpha RNA (group 1, N=72 and group 2, N=73) or protein expression (group 3, N=90). In group 1, patients did not receive any adjuvant treatment, whereas, in groups 2 and 3, patients were treated with anti-estrogens and/or with chemotherapeutical drugs. Rho-GDIalpha RNA levels, measured by RT-PCR from fresh-frozen material, did not correlate with relapse-free survival in Kaplan-Meier analysis, except in a subgroup of CMF-only treated patients. In this subgroup, higher Rho-GDIalpha RNA levels were significantly associated with more favorable prognosis. Immunohistochemical analysis (group 3) confirmed the link between higher Rho-GDIalpha expression and better outcome. This was again particularly true for the CMF-only treated patients. Cox regression analysis revealed that high Rho-GDIalpha protein expression reduced the risk for a relapse by approximately 3-fold, even if adjusted for grading, tumor size, nodal and estrogen receptor (ER) status. The data suggest that Rho-GDIalpha is beneficial to patients who received adjuvant chemotherapy. Rho-GDIalpha is possibly a useful biomarker to predict the response of breast cancer patients to CMF treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/análise , Neoplasias da Mama/tratamento farmacológico , Carcinoma Ductal de Mama/tratamento farmacológico , Inibidores de Dissociação do Nucleotídeo Guanina/biossíntese , Adulto , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Quimioterapia Adjuvante , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fluoruracila/uso terapêutico , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Risco , Resultado do Tratamento , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
13.
Cell Mol Life Sci ; 66(18): 3053-65, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19603142

RESUMO

Mesenchymal stem cells (MSCs) have been shown to communicate with tumor cells. We analyzed the effect of human MSCs (hMSCs) on breast cancer cells in three-dimensional cultures. By using GFP expression and immunohistochemistry, we show that hMSCs invade 3D breast cancer cell aggregates. hMSCs caused breast cancer spheroids to become disorganized which was accompanied by a disruption of cell-cell adhesion, E-cadherin cleavage, and nuclear translocation of E-cadherin, but not by epithelial/mesenchymal transition or by an increase in ERK1/2 activity. In addition, hMSCs enhanced the motility of breast cancer cells. Inhibition of ADAM10 (a disintegrin and metalloprotease 10), known to cleave E-cadherin, prevented both hMSC-mediated E-cadherin cleavage and enhanced migration. Our data suggest that hMSCs interfere with cell-cell adhesion and enhance migration of breast cancer cells by activating ADAM10.


Assuntos
Proteínas ADAM/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Neoplasias da Mama/patologia , Caderinas/metabolismo , Proteínas de Membrana/fisiologia , Células-Tronco Mesenquimais/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Transporte Ativo do Núcleo Celular , Secretases da Proteína Precursora do Amiloide/metabolismo , Adesão Celular , Movimento Celular , Técnicas de Cocultura , Humanos , Proteínas de Membrana/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Células Tumorais Cultivadas
14.
Cancer Lett ; 260(1-2): 56-61, 2008 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-18035481

RESUMO

Parathyroid hormone-related protein (PTHrP) regulates growth and migration of adherent breast cancer cells. Here, we show that PTHrP also interferes with the ability of breast cancer cells to aggregate in suspension cultures. Cell colonies were significantly smaller when the expression of PTHrP or its target genes, integrin alpha6 or KISS-1, was suppressed by RNA interference. TGFbeta1, a stimulator of PTHrP transcription, abolished the effect of PTHrP and KISS-1 specific siRNAs and increased ERK1/2 phosphorylation, whereas inhibition of ERK1/2 phosphorylation by U0126 reduced colony size. PTHrP and KISS-1 may regulate colony formation in 3D by influencing ERK1/2 phosphorylation.


Assuntos
Neoplasias da Mama/metabolismo , Agregação Celular , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Butadienos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Integrina alfa6/genética , Integrina alfa6/metabolismo , Kisspeptinas , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Proteína Relacionada ao Hormônio Paratireóideo/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Esferoides Celulares , Transfecção , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
15.
Cancer Res ; 67(22): 10694-702, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006811

RESUMO

Rho GDP dissociation inhibitor beta (Rho-GDI beta), an inhibitor of Rho GTPases, is primarily expressed by hematopoietic cells but is also found in epithelial cancer cells. Recently, we have identified Rho-GDI beta as a target of the transcription factor Ets1. Here, we show that, in breast cancer cells, Ets1 regulates Rho-GDI beta expression and binds to the upstream region of the Rho-GDI beta gene. Furthermore, in primary breast cancer, Rho-GDI beta is coexpressed with Ets1. Studying the function of Rho-GDI beta in breast cancer, we found that a Rho-GD beta-specific small interfering RNA increased cellular migration but also decreased the expression of cyclooxygenase-2 (Cox-2) oncogene as shown by microarray, quantitative reverse transcription-PCR, and Western blot analyses. Further studies revealed that Rho-GDI beta regulates Cox-2 gene at least partly on the transcriptional level, most likely by activating nuclear factor of activated T cells 1 (NFAT-1). Vav-1, an interaction partner of Rho-GDI beta, was also found to interfere with Cox-2 expression and NFAT-1 cellular distribution, suggesting a cooperative action of Rho-GDI beta and Vav-1 on Cox-2 expression. To explore the importance of Rho-GDI beta for the survival of breast cancer patients, two cohorts, including 263 and 117 patients, were analyzed for clinical outcome in relation to Rho-GDI beta RNA and protein levels, respectively. Expression of Rho-GDI beta was not associated with either disease-free or overall survival in the two patient population. Our data suggest that the expression of Rho-GDI beta in breast cancer is neither beneficial nor disadvantageous to the patient. This may be the net effect of two opposing activities of Rho-GDI beta, one that suppresses tumor progression by inhibiting migration and the other that stimulates it by enhancing Cox-2 expression.


Assuntos
Neoplasias da Mama/metabolismo , Ciclo-Oxigenase 2/fisiologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Biópsia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/mortalidade , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Ciclo-Oxigenase 2/biossíntese , Progressão da Doença , Regulação Enzimológica da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Proteína Proto-Oncogênica c-ets-1/biossíntese , Resultado do Tratamento , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
16.
Electrophoresis ; 27(14): 2844-5, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16688701

RESUMO

Beta-actin is often used as a loading control in Western blot analysis. We analyzed the ability of beta-actin-specific antibodies to recognize differences in protein loading. We found that, at higher total protein loads as required for the detection of low-abundance proteins, beta-actin-specific antibodies failed to distinguish differences in actin protein levels. Diluting the antibody working solution or changing the incubation time had little effect on this phenomenon. This shows that beta-Actin is not a reliable loading control in Western blot analysis. In general, it appeared that, at longer incubation times, antibodies seem to be less able to pick up differences in the level of its target protein.


Assuntos
Actinas/análise , Actinas/imunologia , Western Blotting/normas , Anticorpos/imunologia , Humanos , Proteínas de Neoplasias/análise , Padrões de Referência , Reprodutibilidade dos Testes
17.
J Biol Chem ; 281(21): 14563-72, 2006 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-16551631

RESUMO

The effect of endogenous parathyroid hormone-related protein (PTHrP) on gene expression in breast cancer cells was studied. We suppressed PTHrP expression in MDA-MB-231 cells by RNA interference and analyzed changes in gene expression by microarray analysis. More than 200 genes showed altered expression in response to a PTHrP-specific small interfering (si) RNA (siPTHrP). Cell cycle-regulating gene CDC2 and genes (CDC25B and Tome-1) that control CDC2 activity showed increased expression in the presence of siPTHrP. CDC2 activity was also found to be higher in siPTHrP-treated cells. Studies with PTHrP peptides 1-34 and 67-86, forskolin, and a PTH1 receptor (PTH1R)-specific siRNA showed that PTHrP regulates CDC2 and CDC25B, at least in part, via PTH1R in a cAMP-independent manner. Other siPTHrP-responsive genes included integrin alpha6 (ITGA6), KISS-1, and PAI-1. When combined, siRNAs against ITGA6, PAI-1, and KISS-1 could mimic the negative effect of siPTHrP on migration, whereas siKISS-1 and siPTHrP similarly reduced the proliferative activity of the cells. Comparative expression analyses with 50 primary breast carcinomas revealed that the RNA level of ITGA6 correlates with that of PTHrP, and higher CDC2 and CDC25B values are found at low PTHrP expression. Our data suggest that PTHrP has a profound effect on gene expression in breast cancer cells and, as a consequence, contributes to the regulation of important cellular activities, such as migration and proliferation.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Biópsia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Colforsina/farmacologia , Humanos , Integrina alfa6/metabolismo , Kisspeptinas , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...