Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Steroids ; 200: 109316, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806603

ABSTRACT

Breast cancer is one of the leading causes of cancer-related fatalities and the most often diagnosed malignancy in women globally. Dysregulation of sex hormone signaling pathways mediated by the estrogen receptor (ER) in breast cancer is well characterized. Although ER is known to promote cell growth and survival by altering gene transcription, recent research suggests that its effects in cancers are also mediated through dysregulation of protein synthesis. This implies that ER can coordinately affect gene expression through both translational and transcriptional pathways, leading to the development of malignancy. In this review, we will cover the current understanding of how the ER controls mRNA translation in breast cancer and discuss any potential clinical implications of this phenomenon.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Estrogen Antagonists , Protein Biosynthesis , Signal Transduction/genetics , Gene Expression Regulation, Neoplastic , Estrogen Receptor alpha/metabolism
2.
Chest ; 163(5): 1144-1155, 2023 05.
Article in English | MEDLINE | ID: mdl-36642366

ABSTRACT

BACKGROUND: A critical need exists to develop remission-inducing therapies for lymphangioleiomyomatosis. RESEARCH QUESTION: Is the addition of resveratrol safe and more efficacious than sirolimus alone in patients with lymphangioleiomyomatosis? STUDY DESIGN AND METHODS: We conducted a phase 2, dose-escalating, open-label trial of resveratrol in patients with lymphangioleiomyomatosis receiving a stable regimen of sirolimus. Resveratrol was started at 250 mg/d and escalated every 8 weeks to maximum dose of 1,000 mg/d over 24 weeks. The primary outcome was ≥ 42% decline in serum vascular endothelial growth factor D (VEGF-D) levels on combined therapy compared with baseline VEGF-D levels on sirolimus. Secondary objectives included an assessment of the safety profile and the effect on lung function and health-related quality of life (HRQOL). Longitudinal change in outcome measures was assessed using linear mixed models. Adverse effects were tabulated using the National Cancer Institute's Common Terminology Criteria for Adverse Events version 4. RESULTS: Twenty-five patients with lymphangioleiomyomatosis with a median age of 51 years were enrolled. Pulmonary function parameters at study inclusion were: FEV1: median absolute, 1.72 L; 64% predicted; FVC: median absolute, 2.99 L; 96% predicted; and diffusing capacity of the lungs for carbon monoxide: median absolute, 14.68 mL/mm Hg/min; 37% predicted. The median serum VEGF-D value at baseline was 617 pg/mL. Patients entered the study with a median sirolimus dose of 2 mg/d with median trough level of 6.3 ng/mL. Despite some GI side effects, the addition of resveratrol was well tolerated. Although the primary outcome was not met, a statistically significant reduction in serum VEGF-D levels and improvement in HRQOL during the study was found. INTERPRETATION: The addition of resveratrol was safe and well tolerated in patients with lymphangioleiomyomatosis taking sirolimus and was associated with modest improvement in HRQOL. Larger controlled trials of this combination might be warranted to assess definitively the usefulness of resveratrol as an additive therapy in lymphangioleiomyomatosis. TRIAL REGISTRY: ClinicalTrials.gov; No.: NCT03253913; URL: www. CLINICALTRIALS: gov.


Subject(s)
Lymphangioleiomyomatosis , Sirolimus , Humans , Middle Aged , Sirolimus/therapeutic use , Lymphangioleiomyomatosis/complications , Vascular Endothelial Growth Factor D/metabolism , Resveratrol/therapeutic use , Quality of Life , Forced Expiratory Volume
3.
Am J Physiol Cell Physiol ; 324(2): C320-C326, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36571446

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare disease affecting women, caused by somatic mutations in the TSC1 or TSC2 genes, and driven by estrogen. Similar to many cancers, it is metastatic, primarily to the lung. Despite its monogenetic nature, like many cancers, LAM is a heterogeneous disease. The cellular constituents of LAM are very diverse, including mesenchymal, epithelial, endothelial, and immune cells. LAM is characterized by dysregulation of many cell signaling pathways, distinct populations of LAM cells, and a rich microenvironment, in which the immune system appears to play an important role. This review delineates the heterogeneity of LAM and focuses on the metastatic features of LAM, the deregulated signaling mechanisms and the tumor microenvironment. Understanding the tumor-host interaction in LAM may provide insights into the development of new therapeutic strategies, which could be combinatorial or superlative to Sirolimus, the current U.S. Food and Drug Administration-approved treatment.


Subject(s)
Lung Neoplasms , Lymphangioleiomyomatosis , Humans , Female , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Lymphangioleiomyomatosis/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Tumor Microenvironment
5.
FEBS Lett ; 595(5): 595-603, 2021 03.
Article in English | MEDLINE | ID: mdl-33423298

ABSTRACT

We have previously demonstrated that Fanconi anemia (FA) proteins work in concert with other FA and non-FA proteins to mediate stalled replication fork restart. Previous studies suggest a connection between the FA protein FANCD2 and the non-FA protein mechanistic target of rapamycin (mTOR). A recent study showed that mTOR is involved in actin-dependent DNA replication fork restart, suggesting possible roles in the FA DNA repair pathway. In this study, we demonstrate that during replication stress mTOR interacts and cooperates with FANCD2 to provide cellular stability, mediate stalled replication fork restart, and prevent nucleolytic degradation of the nascent DNA strands. Taken together, this study unravels a novel functional cross-talk between two important mechanisms: mTOR and FA DNA repair pathways that ensure genomic stability.


Subject(s)
DNA Repair/drug effects , DNA Replication/drug effects , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia/genetics , Fibroblasts/metabolism , TOR Serine-Threonine Kinases/genetics , Aphidicolin/pharmacology , Cell Survival/drug effects , DNA/genetics , DNA/metabolism , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fibroblasts/drug effects , Fibroblasts/pathology , Genome, Human , Genomic Instability , Humans , Hydroxyurea/pharmacology , Mitomycin/pharmacology , Primary Cell Culture , Protein Binding/drug effects , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
6.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118907, 2021 02.
Article in English | MEDLINE | ID: mdl-33189783

ABSTRACT

Positioned at the axis between the cell and its environment, mTOR directs a wide range of cellular activity in response to nutrients, growth factors, and stress. Our understanding of the role of mTOR is evolving beyond the spatial confines of the cytosol, and its role in the nucleus becoming ever more apparent. In this review, we will address various studies that explore the role of nuclear mTOR (nmTOR) in specific cellular programs and how these pathways influence one another. To understand the emerging roles of nuclear mTOR, we discuss data and propose plausible mechanisms to offer novel ideas, hypotheses, and future research directions.


Subject(s)
Cell Nucleus/metabolism , TOR Serine-Threonine Kinases/metabolism , Humans
7.
Endocrinology ; 161(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-32128594

ABSTRACT

Estrogen is a key hormone involved in the development and homeostasis of several tissue types in both males and females. By binding estrogen receptors, estrogen regulates essential functions of gene expression, metabolism, cell growth, and proliferation by acting through cytoplasmic signaling pathways or activating transcription in the nucleus. However, disruption or dysregulation of estrogen activity has been shown to play a key role in the pathogenesis and progression of many diseases. This review will expatiate on some of the unconventional roles of estrogen in homeostasis and disease.


Subject(s)
Cell Membrane/metabolism , Estrogens/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans
8.
Mol Cell Proteomics ; 18(8): 1607-1618, 2019 08.
Article in English | MEDLINE | ID: mdl-31189691

ABSTRACT

ER-positive breast tumors represent ∼70% of all breast cancer cases. Although their treatment with endocrine therapies is effective in the adjuvant or recurrent settings, the development of resistance compromises their effectiveness. The binding of estrogen to ERα, a transcription factor, triggers the regulation of the target genes (genomic pathway). Additionally, a cytoplasmic fraction of estrogen-bound ERα activates oncogenic signaling pathways such as PI3K/AKT/mTOR (nongenomic pathway). The upregulation of the estrogenic and the PI3K/AKT/mTOR signaling pathways are frequently associated with a poor outcome. To better characterize the connection between these two pathways, we performed a phosphoproteome analysis of ER-positive MCF7 breast cancer cells treated with estrogen or estrogen and the mTORC1 inhibitor rapamycin. Many proteins were identified as estrogen-regulated mTORC1 targets and among them, DEPTOR was selected for further characterization. DEPTOR binds to mTOR and inhibits the kinase activity of both mTOR complexes mTORC1 and mTORC2, but mitogen-activated mTOR promotes phosphorylation-mediated DEPTOR degradation. Although estrogen enhances the phosphorylation of DEPTOR by mTORC1, DEPTOR levels increase in estrogen-stimulated cells. We demonstrated that DEPTOR accumulation is the result of estrogen-ERα-mediated transcriptional upregulation of DEPTOR expression. Consequently, the elevated levels of DEPTOR partially counterbalance the estrogen-induced activation of mTORC1 and mTORC2. These results underscore the critical role of estrogen-ERα as a modulator of the PI3K/AKT/mTOR signaling pathway in ER-positive breast cancer cells. Additionally, these studies provide evidence supporting the use of dual PI3K/mTOR or dual mTORC1/2 inhibitors in combination with endocrine therapies as a first-line treatment option for the patients with ER-positive advanced breast cancer.


Subject(s)
Estrogen Receptor alpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Estrogens/pharmacology , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Phosphorylation , Proteome , Sirolimus/pharmacology
9.
J Biol Chem ; 294(7): 2267-2278, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30573685

ABSTRACT

Approximately two thirds of all breast cancer cases are estrogen receptor (ER)-positive. The treatment of this breast cancer subtype with endocrine therapies is effective in the adjuvant and recurrent settings. However, their effectiveness is compromised by the emergence of intrinsic or acquired resistance. Thus, identification of new molecular targets can significantly contribute to the development of novel therapeutic strategies. In recent years, many studies have implicated aberrant levels of translation initiation factors in cancer etiology and provided evidence that identifies these factors as promising therapeutic targets. Accordingly, we observed reduced levels of the eIF3 subunit eIF3f in ER-positive breast cancer cells compared with ER-negative cells, and determined that low eIF3f levels are required for proper proliferation and survival of ER-positive MCF7 cells. The expression of eIF3f is tightly controlled by ERα at the transcriptional (genomic pathway) and translational (nongenomic pathway) level. Specifically, estrogen-bound ERα represses transcription of the EIF3F gene, while promoting eIF3f mRNA translation. To regulate translation, estrogen activates the mTORC1 pathway, which enhances the binding of eIF3 to the eIF4F complex and, consequently, the assembly of the 48S preinitiation complexes and protein synthesis. We observed preferential translation of mRNAs with highly structured 5'-UTRs that usually encode factors involved in cell proliferation and survival (e.g. cyclin D1 and survivin). Our results underscore the importance of estrogen-ERα-mediated control of eIF3f expression for the proliferation and survival of ER-positive breast cancer cells. These findings may provide rationale for the development of new therapies to treat ER-positive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Eukaryotic Initiation Factor-3/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Signal Transduction , 3' Untranslated Regions , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Survival , Estrogen Receptor alpha/genetics , Eukaryotic Initiation Factor-3/genetics , Female , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasm Proteins/genetics
10.
Breast Cancer Res Treat ; 168(1): 17-27, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29128895

ABSTRACT

PURPOSE: Deregulated PI3K/mTOR signals can promote the growth of breast cancer and contribute to endocrine treatment resistance. This report aims to investigate raptor and its intracellular localization to further understand its role in ER-positive breast cancer. METHODS: Raptor protein expression was evaluated by immunohistochemistry in 756 primary breast tumors from postmenopausal patients randomized to tamoxifen or no tamoxifen. In vitro, the MCF7 breast cancer cell line and tamoxifen-resistant MCF7 cells were studied to track the raptor signaling changes upon resistance, and raptor localization in ERα-positive cell lines was compared with that in ERα-negative cell lines. RESULTS: Raptor protein expression in the nucleus was high in ER/PgR-positive and HER2-negative tumors with low grade, features associated with the luminal A subtype. Presence of raptor in the nucleus was connected with ERα signaling, here shown by a coupled increase of ERα phosphorylation at S167 and S305 with accumulation of nuclear raptor. In addition, the expression of ERα-activated gene products correlated with nuclear raptor. Similarly, in vitro we observed raptor in the nucleus of ERα-positive, but not of ER-negative cells. Interestingly, raptor localized to the nucleus could still be seen in tamoxifen-resistant MCF7 cells. The clinical benefit from tamoxifen was inversely associated with an increase of nuclear raptor. High cytoplasmic raptor expression indicated worse prognosis on long-term follow-up. CONCLUSION: We present a connection between raptor localization to the nucleus and ERα-positive breast cancer, suggesting raptor as a player in stimulating the growth of the luminal A subtype and a possible target along with endocrine treatment.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast/cytology , Breast/pathology , Breast/surgery , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Cell Line, Tumor , Chemotherapy, Adjuvant/methods , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mastectomy , Phosphorylation , Postmenopause , Prognosis , Randomized Controlled Trials as Topic , Receptors, Progesterone/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tamoxifen/therapeutic use , Treatment Outcome
11.
Article in English | MEDLINE | ID: mdl-28989978

ABSTRACT

Resveratrol is a nutraceutical with several therapeutic effects. It has been shown to mimic effects of caloric restriction, exert anti-inflammatory and anti-oxidative effects, and affect the initiation and progression of many diseases through several mechanisms. While there is a wealth of in vitro and in vivo evidence that resveratrol could be a promising therapeutic agent, clinical trials must confirm its potential. In this work, we reviewed the current clinical data available regarding the pharmacological action of resveratrol. Most of the clinical trials of resveratrol have focused on cancer, neurological disorders, cardiovascular diseases, diabetes, non-alcoholic fatty liver disease (NAFLD), and obesity. We found that for neurological disorders, cardiovascular diseases, and diabetes, the current clinical trials show that resveratrol was well tolerated and beneficially influenced disease biomarkers. However resveratrol had ambiguous and sometimes even detrimental effects in certain types of cancers and in NAFLD. In most of the clinical trials, the major obstacle presented was resveratrol's poor bioavailability. Thus, this work provides useful considerations for the planning and design of future pre-clinical and clinical research on resveratrol.

12.
Article in English | MEDLINE | ID: mdl-28890840

ABSTRACT

Estrogen-related receptor alpha (ERRα) is an orphan nuclear factor that is a master regulator of cellular energy metabolism. ERRα is overexpressed in a variety of tumors, including ovarian, prostate, colorectal, cervical and breast, and is associated with a more aggressive tumor and a worse outcome. In breast cancer, specifically, high ERRα expression is associated with an increased rate of recurrence and a poor prognosis. Because of the common functions of ERRα and the mTORC1/S6K1 signaling pathway in regulation of cellular metabolism and breast cancer pathogenesis, we focused on investigating the biochemical relationship between ERRα and S6K1. We found that ERRα negatively regulates S6K1 expression by directly binding to its promoter. Downregulation of ERRα expression sensitized ERα-negative breast cancer cells to mTORC1/S6K1 inhibitors. Therefore, our results show that combinatorial inhibition of ERRα and mTORC1/S6K1 may have clinical utility in treatment of triple-negative breast cancer, and warrants further investigation.

13.
Mol Cancer Res ; 15(10): 1318-1330, 2017 10.
Article in English | MEDLINE | ID: mdl-28710231

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor-suppressor syndrome affecting multiple organs, including the brain, skin, kidneys, heart, and lungs. TSC is associated with mutations in TSC1 or TSC2, resulting in hyperactivation of mTOR complex 1 (mTORC1). Clinical trials demonstrate that mTORC1 inhibitors decrease tumor volume and stabilize lung function in TSC patients; however, mTOR inhibitors are cytostatic not cytocidal, and long-term benefits and toxicities are uncertain. Previously, we identified rapamycin-insensitive upregulation of cyclooxygenase 2 (PTGS2/COX2) and prostaglandin E2 (PGE2) production in TSC2-deficient cells and postulated that the action of excess PGE2 and its cognate receptors (EP) contributes to cell survival. In this study, we identify upregulation of EP3 (PTGER3) expression in TSC2-deficient cells, TSC renal angiomyolipomas, lymphangioleiomyomatosis lung nodules, and epileptic brain tubers. TSC2 negatively regulated EP3 expression via Rheb in a rapamycin-insensitive manner. The EP3 antagonist, L-798106, selectively suppressed the viability of TSC2-deficient cells in vitro and decreased the lung colonization of TSC2-deficient cells. Collectively, these data reveal a novel function of TSC2 and Rheb in the regulation of EP3 expression and cell viability.Implications: Therapeutic targeting of an aberrant PGE2-EP3 signaling axis may have therapeutic benefit for TSC patients and for other mTOR-hyperactive neoplasms. Mol Cancer Res; 15(10); 1318-30. ©2017 AACR.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Tumor Suppressor Proteins/metabolism , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Child , Child, Preschool , Epilepsy/genetics , Epilepsy/metabolism , Female , Humans , Infant , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Male , Mice , Mutation , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Up-Regulation
14.
J Cell Physiol ; 232(2): 436-446, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27225870

ABSTRACT

Loss of TSC1 function, a crucial negative regulator of mTOR signaling, is a common alteration in bladder cancer. Mutations in other members of the PI3K pathway, leading to mTOR activation, are also found in bladder cancer. This provides rationale for targeting mTOR for treatment of bladder cancer characterized by TSC1 mutations and/or mTOR activation. In this study, we asked whether combination treatment with rapamycin and resveratrol could be effective in concurrently inhibiting mTOR and PI3K signaling and inducing cell death in bladder cancer cells. In combination with rapamycin, resveratrol was able to block rapamycin-induced Akt activation, while maintaining mTOR pathway inhibition. In addition, combination treatment with rapamycin and resveratrol induced cell death specifically in TSC1-/- MEF cells, and not in wild-type MEFs. Similarly, resveratrol alone or in combination with rapamycin induced cell death in human bladder cancer cell lines. These data indicate that administration of resveratrol together with rapamycin may be a promising therapeutic option for treatment of bladder cancer. J. Cell. Physiol. 232: 436-446, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Sirolimus/therapeutic use , Stilbenes/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Fibroblasts/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Resveratrol , Signal Transduction/drug effects , Sirolimus/pharmacology , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology
15.
Cell Cycle ; 15(23): 3230-3239, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27753535

ABSTRACT

Homologous recombination (HR) is a conserved process that maintains genome stability and cell survival by repairing DNA double-strand breaks (DSBs). The RAD51-related family of proteins is involved in repair of DSBs; consequently, deregulation of RAD51 causes chromosomal rearrangements and stimulates tumorigenesis. RAD51C has been identified as a potential tumor suppressor and a breast and ovarian cancer susceptibility gene. Recent studies have also implicated estrogen as a DNA-damaging agent that causes DSBs. We found that in ERα-positive breast cancer cells, estrogen transcriptionally regulates RAD51C expression in ERα-dependent mechanism. Moreover, estrogen induces RAD51C assembly into nuclear foci at DSBs, which is a precursor to RAD51 complex recruitment to the nucleus. Additionally, disruption of ERα signaling by either anti-estrogens or siRNA prevented estrogen induced upregulation of RAD51C. We have also found an association of a worse clinical outcome between RAD51C expression and ERα status of tumors. These findings provide insight into the mechanism of genomic instability in ERα-positive breast cancer and suggest that individuals with mutations in RAD51C that are exposed to estrogen would be more susceptible to accumulation of DNA damage, leading to cancer progression.


Subject(s)
DNA Damage/genetics , DNA-Binding Proteins/genetics , Estrogens/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Databases, Genetic , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Transport/drug effects , Transcription, Genetic/drug effects , Treatment Outcome
16.
Oncotarget ; 7(19): 27567-83, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27028868

ABSTRACT

The p90 ribosomal S6 kinase (RSK) is a family of MAPK-activated serine/threonine kinases (RSK1-4) whose expression and/or activity are deregulated in several cancers, including breast cancer. Up-regulation of RSKs promotes cellular processes that drive tumorigenesis in Triple Negative Breast Cancer (TNBC) cells. Although RSKs regulate protein synthesis in certain cell types, the role of RSK-mediated translational control in oncogenic progression has yet to be evaluated. We demonstrate that proliferation and migration of TNBC MDA-MB-231 cells, unlike ER/PR-positive MCF7 cells, rely on RSK activity. We show that RSKs regulate the activities of the translation initiation factor eIF4B and the translational repressor PDCD4 in TNBC cells with up-regulated MAPK pathway, but not in breast cancer cells with hyperactivated PI3K/Akt/mTORC1 pathway. These results identify PDCD4 as a novel RSK substrate. We demonstrate that RSK-mediated phosphorylation of PDCD4 at S76 promotes PDCD4 degradation. Low PDCD4 levels reduce PDCD4 inhibitory effect on the translation initiation factor eIF4A, which increases translation of "eIF4A sensitive" mRNAs encoding factors involved in cell cycle progression, survival, and migration. Consequently, low levels of PDCD4 favor proliferation and migration of MDA-MB-231 cells. These results support the therapeutic use of RSK inhibitors for treatment of TNBC with deregulated MAPK/RSK pathway.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , HEK293 Cells , Humans , MCF-7 Cells , RNA-Binding Proteins/genetics , Signal Transduction , Transfection , Triple Negative Breast Neoplasms/genetics
17.
Sign Transduct Insights ; 5: 1-7, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26989346

ABSTRACT

Breast cancer is a highly heterogeneous disease. Tamoxifen is a selective estrogen receptor (ER) modulator and is mainly indicated for the treatment of breast cancer in postmenopausal women and postsurgery neoadjuvant therapy in ER-positive breast cancers. Interestingly, 5-10% of the ER-negative breast cancers have also shown sensitivity to tamoxifen treatment. The involvement of molecular markers and/or signaling pathways independent of ER signaling has been implicated in tamoxifen sensitivity in the ER-negative subgroup. Studies reveal that variation in the expression of estrogen-related receptor alpha, ER subtype beta, tumor microenvironment, and epigenetics affects tamoxifen sensitivity. This review discusses the background of the research on the action of tamoxifen that may inspire future studies to explore effective therapeutic strategies for the treatment of ER-negative and triple-negative breast cancers, the latter being an aggressive disease with worse clinical outcome.

18.
Clin Cancer Res ; 22(6): 1421-31, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26542058

ABSTRACT

PURPOSE: Estrogen-related receptor alpha (ERRα) signaling has recently been implicated in breast cancer. We investigated the clinical value of ERRα in randomized cohorts of tamoxifen-treated and adjuvant-untreated patients. EXPERIMENTAL DESIGN: Cox proportional hazards regression was used to evaluate the significance of associations between ERRα gene expression levels and patient DMFS in a previously published microarray dataset representing 2,000 breast tumor cases derived from multiple medical centers worldwide. The 912 tumors used for immunostaining were from a tamoxifen-randomized primary breast cancer trial conducted in Stockholm, Sweden, during 1976-1990. Mouse model was used to study the effect of tamoxifen treatment on lung colonization of MDA-MB-231 control cells and MDA-MB-231 cells with stable knockdown of ERRα. The phenotypic effects associated with ERRα modulation were studied using immunoblotting analyses and wound-healing assay. RESULTS: We found that in ER-negative and triple-negative breast cancer (TNBC) adjuvant-untreated patients, ERRα expression indicated worse prognosis and correlated with poor outcome predictors. However, in tamoxifen-treated patients, an improved outcome was observed with high ERRα gene and protein expression. Reduced ERRα expression was oncogenic in the presence of tamoxifen, measured by in vitro proliferation and migration assays and in vivo metastasis studies. CONCLUSIONS: Taken together, these data show that ERRα expression predicts response to tamoxifen treatment, and ERRα could be a biomarker of tamoxifen sensitivity and a prognostic factor in TNBC.


Subject(s)
Biomarkers, Tumor , Receptors, Estrogen/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , Female , Gene Expression , Gene Knockout Techniques , Humans , Neoplasm Grading , Prognosis , Protein Transport , RNA, Small Interfering/genetics , Receptors, Estrogen/genetics , Tamoxifen/therapeutic use , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , ERRalpha Estrogen-Related Receptor
19.
Ann N Y Acad Sci ; 1348(1): 116-23, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26200935

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is hyperactivated in a variety of cancers and tumor syndromes. Therefore, mTORC1 inhibitors are being actively investigated for treatment of neoplasms. The concern with the monotherapy use of mTORC1 inhibitors, such as rapamycin, is that they cause upregulation of autophagy, a cell survival mechanism, and suppress the negative feedback loop to the oncogene Akt. In turn, Akt promotes cell survival, causing the therapy to be partially effective, but relapse occurs upon cessation of treatment. In this review, we describe the current literature on resveratrol as well as our work, which uses rapamycin in combination with resveratrol. We found that this combination treatment efficiently blocked upregulation of autophagy and restored inhibition of Akt in different cancer and tumor models. Interestingly, the combination of rapamycin and resveratrol selectively promoted apoptosis of cells with mTOR pathway hyperactivation. Moreover, this combination prevented tumor growth and lung metastasis when tested in mouse models. Finally, mass spectrometry-based identification of cellular targets of resveratrol provided mechanistic insight into the mode of action of resveratrol. The addition of resveratrol to rapamycin treatment may be a promising option for selective and targeted therapy for diseases with mTORC1 hyperactivation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/metabolism , Signal Transduction , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Resveratrol , Stilbenes/therapeutic use , Xenograft Model Antitumor Assays
20.
Am J Respir Cell Mol Biol ; 53(5): 637-46, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25844891

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare neoplastic metastatic disease affecting women of childbearing age. LAM is caused by hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) as a consequence of tuberous sclerosis complex (TSC) 1/2 inactivation. Clinically, LAM results in cystic lung destruction. mTORC1 inhibition using rapamycin analogs (rapalogs) is partially effective in reducing disease progression and improving lung function. However, cessation of treatment results in continued progression of the disease. In the present study, we investigated the effectiveness of the combination of rapamycin treatment with resveratrol, an autophagy inhibitor, in the TSC2-null xenograft tumor model. We determined that this combination inhibits phosphatidylinositol-4,5-bisphosphate 3-kinase PI3K/Akt/mTORC1 signaling and activates apoptosis. Therefore, the combination of rapamycin and resveratrol may be an effective clinical strategy for treatment of LAM and other diseases with mTORC1 hyperactivation.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Lymphangioleiomyomatosis/drug therapy , Sirolimus/pharmacology , Stilbenes/pharmacology , Tumor Suppressor Proteins/genetics , Uterine Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Drug Therapy, Combination , Female , Humans , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/immunology , Lymphangioleiomyomatosis/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice, SCID , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Rats , Resveratrol , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Treatment Outcome , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/immunology , Uterine Neoplasms/genetics , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...