Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Gynecol Oncol ; 177: 60-71, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37639904

ABSTRACT

OBJECTIVE: ATR kinase inhibitors promote cell killing by inducing replication stress and through potentiation of genotoxic agents in gynecologic cancer cells. To explore mechanisms of acquired resistance to ATRi in ovarian cancer, we characterized ATRi-resistant ovarian cancer cells generated by metronomic dosing with the clinical ATR inhibitor AZD6738. METHODS: ATRi-resistant ovarian cancer cells (OVCAR3 and OV90) were generated by dosing with AZD6738 and assessed for sensitivity to Chk1i (LY2603618), PARPi (Olaparib) and combination with cisplatin or a CDK4/6 inhibitor (Palbociclib). Models were characterized by diverse methods including silencing CDC25A in OV90 cells and assessing impact on ATRi response. Serum proteomic analysis of ATRi-resistant OV90 xenografts was performed to identify circulating biomarker candidates of ATRi-resistance. RESULTS: AZD6738-resistant cell lines are refractory to LY2603618, but not to Olaparib or combinations with cisplatin. Cell cycle analyses showed ATRi-resistant cells exhibit G1/S arrest following AZD6738 treatment. Accordingly, combination with Palbociclib confers resistance to AZD6738. AZD6738-resistant cells exhibit altered abundances of G1/S phase regulatory proteins, including loss of CDC25A in AZD6738-resistant OV90 cells. Silencing of CDC25A in OV90 cells confers resistance to AZD6738. Serum proteomics from AZD6738-resistant OV90 xenografts identified Vitamin D-Binding Protein (GC), Apolipoprotein E (APOE) and A1 (APOA1) as significantly elevated in AZD6738-resistant backgrounds. CONCLUSIONS: We show that metronomic dosing of ovarian cancer cells with AZD6738 results in resistance to ATR/ Chk1 inhibitors, that loss of CDC25A expression represents a mechanism of resistance to ATRi treatment in ovarian cancer cells and identify several circulating biomarker candidates of CDC25A low, AZD6738-resistant ovarian cancer cells.

2.
J Drug Target ; 30(6): 623-633, 2022 07.
Article in English | MEDLINE | ID: mdl-35100927

ABSTRACT

Endometrial cancer (EC) is a common and deadly cancer in women and novel therapeutic approaches are urgently needed. Polyamines (putrescine, spermidine, spermine) are critical for mammalian cell proliferation and MYC coordinately regulates polyamine metabolism through ornithine decarboxylase (ODC). ODC is a MYC target gene and rate-limiting enzyme of polyamine biosynthesis and the FDA-approved anti-protozoan drug α-difluoromethylornithine (DFMO) inhibits ODC activity and induces polyamine depletion that leads to tumour growth arrest. Spermidine is required for the hypusine-dependent activation of eukaryotic translation initiation factors 5A1 (eIF5A1) and 5A2 (eIF5A2) and connects the MYC/ODC-induced deregulation of spermidine to eIF5A1/2 protein translation, which is increased during cancer cell proliferation. We show that eIF5A1 is significantly upregulated in EC cells compared to control cells (p=.000038) and that combined pharmacological targeting of ODC and eIF5A hypusination with cytostatic drugs DFMO and N1-guanyl-1,7-diaminoheptane (GC7), respectively, reduces eIF5A1 activation and synergistically induces apoptosis in EC cells. In vivo, DFMO/GC7 suppressed xenografted EC tumour growth in mice more potently than each drug alone compared to control (p=.002) and decreased putrescine (p=.045) and spermidine levels in tumour tissues. Our data suggest DFMO and GC7 combination therapy may be useful in the treatment or prevention of EC.


Subject(s)
Endometrial Neoplasms , Polyamines , Animals , Eflornithine/pharmacology , Endometrial Neoplasms/drug therapy , Female , Humans , Lysine/analogs & derivatives , Mammals/metabolism , Mice , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , Putrescine/metabolism , Spermidine/metabolism , Spermidine/pharmacology , Spermine/metabolism , Spermine/pharmacology
3.
Biochem Biophys Res Commun ; 550: 151-157, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33706098

ABSTRACT

Endometriosis is a disorder in which endometrial cells normally limited to the lining of the uterus proliferate outside the uterine cavity and can cause pelvic pain and infertility. ARID1A levels are significantly reduced in the eutopic endometrium from women with endometriosis. Uterine specific Arid1a knock-out mice were infertile due to loss of epithelial progesterone receptor (PGR) signaling. However, the functional association of ARID1A and PGR in endometriosis has not been studied. We examined the expression patterns and co-localization of ARID1A and PGR in eutopic endometrium from women with and without endometriosis using immunostaining and Western blot analysis. ARID1A and PGR proteins co-localized in the epithelium during the proliferative and the early secretory phases. Our immunoprecipitation analysis and proximity ligation assay (PLA) revealed physical interaction between ARID1A and PGR-A but not PGR-B in the mouse and human endometrium. ARID1A levels positively correlated with PGR levels in the eutopic endometrium of women with endometriosis. Our results bring new perspectives on the molecular mechanisms involved in endometrial receptivity and progesterone resistance in endometriosis. The interrelationship between ARID1A and PGR may contribute to explaining the non-receptive endometrium in endometriosis-related infertility.


Subject(s)
DNA-Binding Proteins/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Receptors, Progesterone/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/deficiency , Endometriosis/pathology , Endometrium/pathology , Female , HEK293 Cells , Humans , Immunoprecipitation , Mice , Protein Binding , Receptors, Progesterone/deficiency , Transcription Factors/deficiency
4.
Heliyon ; 6(5): e04075, 2020 May.
Article in English | MEDLINE | ID: mdl-32490257

ABSTRACT

Endometrial cancer is the most common gynecologic malignancy in the U.S. with metastatic disease remaining the major cause of patient death. Therapeutic strategies have remained essentially unchanged for decades. A significant barrier to progression in treatment modalities stems from a lack of clinically applicable in vivo models to accurately mimic endometrial cancer; specifically, ones that form distant metastases and maintain an intact immune system. To address this problem, we have established the first immune competent murine orthotopic tumor model for metastatic endometrial cancer by creating a green fluorescent protein labeled cell line from an endometrial cancer that developed in a Pgr cre/+ Pten f/f Kras G12D genetically engineered mouse. These cancer cells were grafted into the abraded uterine lumen of ovariectomized recipient mice treated with estrogen and subsequently developed local and metastatic endometrial tumors. We noted primary tumor formation in 59% mixed background and 86% of C57BL/6 animals at 4 weeks and distant lung metastases in 78% of mice after 2 months. This immunocompetent orthotopic tumor model closely resembles some human metastatic endometrial cancer, modeling both local metastasis and hematogenous spread to lung and has significant potential to advance the study of endometrial cancer and its metastasis.

5.
PLoS One ; 12(12): e0189044, 2017.
Article in English | MEDLINE | ID: mdl-29240775

ABSTRACT

Ornithine Decarboxylase (ODC) a key enzyme in polyamine biosynthesis is often overexpressed in cancers and contributes to polyamine-induced cell proliferation. We noted ubiquitous expression of ODC1 in our published endometrial cancer gene array data and confirmed this in the cancer genome atlas (TCGA) with highest expression in non-endometrioid, high grade, and copy number high cancers, which have the worst clinical outcomes. ODC1 expression was associated with worse overall survival and increased recurrence in three endometrial cancer gene expression datasets. Importantly, we confirmed these findings using quantitative real-time polymerase chain reaction (qRT-PCR) in a validation cohort of 60 endometrial cancers and found that endometrial cancers with elevated ODC1 had significantly shorter recurrence-free intervals (KM log-rank p = 0.0312, Wald test p = 5.59e-05). Difluoromethylornithine (DFMO) a specific inhibitor of ODC significantly reduced cell proliferation, cell viability, and colony formation in cell line models derived from undifferentiated, endometrioid, serous, carcinosarcoma (mixed mesodermal tumor; MMT) and clear cell endometrial cancers. DFMO also significantly reduced human endometrial cancer ACI-98 tumor burden in mice compared to controls (p = 0.0023). ODC-regulated polyamines (putrescine [Put] and/or spermidine [Spd]) known activators of cell proliferation were strongly decreased in response to DFMO, in both tumor tissue ([Put] (p = 0.0006), [Spd] (p<0.0001)) and blood plasma ([Put] (p<0.0001), [Spd] (p = 0.0049)) of treated mice. Our study indicates that some endometrial cancers appear particularly sensitive to DFMO and that the polyamine pathway in endometrial cancers in general and specifically those most likely to suffer adverse clinical outcomes could be targeted for effective treatment, chemoprevention or chemoprevention of recurrence.


Subject(s)
Endometrial Neoplasms/drug therapy , Ornithine Decarboxylase/drug effects , Animals , Cohort Studies , Endometrial Neoplasms/enzymology , Female , Humans , Mice , Mice, Nude , Ornithine Decarboxylase/metabolism , Real-Time Polymerase Chain Reaction
6.
Cancer Res ; 74(24): 7371-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25377472

ABSTRACT

PTEN mutations are the most common genetic alterations in endometrial cancer. Loss of PTEN and subsequent AKT activation stimulate estrogen receptor α-dependent pathways that play an important role in endometrial tumorigenesis. The major pathologic phenomenon of endometrial cancer is the loss of ovarian steroid hormone control over uterine epithelial cell proliferation and apoptosis. However, the precise mechanism of PTEN/AKT signaling in endometrial cancer remains poorly understood. The progesterone signaling mediator MIG-6 suppresses estrogen signaling and it has been implicated previously as a tumor suppressor in endometrial cancer. In this study, we show that MIG-6 also acts as a tumor suppressor in endometrial cancers associated with PTEN deficiency. Transgenic mice, where Mig-6 was overexpressed in progesterone receptor-expressing cells, exhibited a relative reduction in uterine tumorigenesis caused by Pten deficiency. ERK1/2 was phosphorylated in uterine tumors and administration of an ERK1/2 inhibitor suppressed cancer progression in PR(cre/+)Pten(f/f) mice. In clinical specimens of endometrial cancer, MIG-6 expression correlated inversely with ERK1/2 phosphorylation during progression. Taken together, our findings suggest that Mig-6 regulates ERK1/2 phosphorylation and that it is crucial for progression of PTEN-mutant endometrial cancers, providing a mechanistic rationale for the evaluation of ERK1/2 inhibitors as a therapeutic treatment in human endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Intracellular Signaling Peptides and Proteins/genetics , PTEN Phosphohydrolase/genetics , Transcriptional Activation , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , MAP Kinase Signaling System/genetics , Mice , Mice, Transgenic , PTEN Phosphohydrolase/deficiency , Phosphorylation
7.
J Cancer Prev ; 18(4): 313-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25337560

ABSTRACT

BACKGROUND: Endometrial cancer, the most common gynecological cancer, is closely associated with endometrial hyperplasia, unopposed estrogen exposure, and genetic alterations. Phosphatase and tensin homologue (PTEN) is a tumor suppressor genes completely lost or mutated in >50% of primary endometrioid endometrial cancers. Estrogen-dependent endometrioid carcinoma is the most common type of endometrial cancer. Progesterone is a hormone that antagonizes the growth-promoting properties of estrogen in the uterus. Progestin is used as a conservative endocrine treatment of early endometrial cancer in order to preserve fertility as well as a palliative measure for advanced-stage patients. Progesterone therapy has been shown to be effective in preventing endometrial cancer as well as controlling growth of the endometrium. However, the effectiveness of progestin for women with endometrial cancer is less clear. METHODS: In order to understand the effect of steroid hormone on endometrial cancer progression, we used a mouse endometrial cancer model with conditional loss of Pten in the mouse uterus (PR (cre/+) Pten (f/f) , Pten(d/d) ). To assess the effect of steroid hormones, ovariectomized Pten(f/f) and Pten(d/d) mice were treated with estrogen or progesterone over a period of three month. RESULTS: Uterine weight gain was significantly decreased in ovariectomized PR (cre/+) Pten(f/f) mice compared to intact PR (cre/+) Pten(f/f) mice. Ovariectomized PR (cre/+) Pten(f/f) mice treated with P4 or vehicle also exhibited decreased uterine cancer size compared with intact PR (cre/+) Pten(f/f) mice. Proliferation of ovariectomized PR (cre/+) Pten(f/f) mice treated with P4 is highly decreased compared to other groups. The levels of stromal progesterone receptor were highly increased in ovariectomized PR (cre/+) Pten(f/f) mice treated with P4 which resulted in decreased epithelial proliferation. CONCLUSIONS: These results suggest that P4 treatment significantly reduces tumor mass but does not affect cancer progression in PR (cre/+) Pten(f/f) mice.

8.
Brain Res ; 1370: 34-42, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21081117

ABSTRACT

Minocycline is an inhibitor of microglial activation and proliferation. Minocycline suppresses pain-related behaviors in many different pain states, which correlates closely with its inhibition of microglial activation and subsequent release of pro-inflammatory mediators in the spinal cord. Na(+) channels in dorsal root ganglion (DRG) neurons are implicated in the generation of inflammatory and neuropathic pain. To elucidate a possible peripheral mechanism of minocycline analgesia, effects of minocycline on tetrodotoxin-sensitive and tetrodotoxin-resistant Na(+) currents in rat DRG neurons were investigated. Minocycline potently inhibited both types of Na(+) currents with IC(50) values of 350 nM and 410 nM, respectively. The inhibition was accompanied by a depolarizing shift of the activation voltage. However, minocycline slowed the inactivation and speeded up the recovery from inactivation. These results suggest minocycline may exert analgesia peripherally thorough Na(+) channel inhibition in the primary afferent neurons as well as centrally through microglial inhibition in the spinal cord.


Subject(s)
Analgesics/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Minocycline/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sodium Channels/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Ganglia, Spinal/cytology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology , Sodium Channels/physiology , Tetrodotoxin/pharmacology
9.
Anticancer Res ; 30(11): 4485-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21115897

ABSTRACT

BACKGROUND: Integrin α6ß4 is a known tumor antigen; however, its function in different subtypes of thyroid cancer is not known. This study reports that α6ß4 expression is selectively up-regulated in anaplastic thyroid cancer (ATC) cells, the most malignant subtype of human thyroid cancer. MATERIALS AND METHODS: To assess the contribution of α6ß4 in ATC progression, cell proliferation, motility and soft agar assay were performed in vitro and a xenograft tumor growth assay was performed in vivo. RESULTS: Knockdown of ß4 integrin subunit expression by shRNA in ATC cells reduced the proliferation, migration, and anchorage-independent growth of ATC cells in vitro and xenograft tumor growth in vivo. CONCLUSION: These data suggest that integrin α6ß4 contributes to the development of aggressive forms of thyroid cancer with poor prognostic potential, such as ATC, and thus may be a novel therapeutic target for the treatment for this subtype of thyroid cancer.


Subject(s)
Carcinoma/genetics , Cell Movement , Cell Proliferation , Integrin alpha6beta4/genetics , RNA Interference , RNA, Small Interfering/genetics , Thyroid Neoplasms/genetics , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Carcinoma/pathology , Cell Adhesion , Cell Line, Tumor , Female , Flow Cytometry , Humans , Integrin alpha6beta4/metabolism , Mice , Mice, Nude , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Thyroid Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Mol Cancer Res ; 8(12): 1571-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21047768

ABSTRACT

It was previously shown that integrin α6ß4 contributes to translation of cancer-related mRNAs such as VEGF via initiation factor eIF4E. In this study, we found that integrin α6ß4 regulates the activity of eIF4E through the Ser/Thr kinase Mnk. Although a role for Mnk in various aspects of cancer progression has been established, a link between integrin and Mnk activity has not. Here we show that Mnk1 is a downstream effector of integrin α6ß4 and mediates the α6ß4 signaling, important for translational control. Integrin α6ß4 signals through MEK and p38 MAPK to increase phosphorylation of Mnk1 and eIF4E. Inhibition of Mnk1 activity by CGP57380 or downregulation by shRNA blocks α6ß4-dependent translation of VEGF mRNA. Our studies suggest that Mnk1 could be a therapeutic target in cancers where the integrin α6ß4 level is high.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Integrin alpha6beta4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , Aniline Compounds/pharmacology , Butadienes/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Culture Media, Serum-Free , Down-Regulation , Eukaryotic Initiation Factor-4E/genetics , Humans , Integrin alpha6beta4/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Nitriles/pharmacology , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Purines/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Biochem Pharmacol ; 80(12): 1921-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20599779

ABSTRACT

Metastasis of primary tumors to distant sites and their inherent or acquired resistance to currently available therapies pose major clinical challenge to the successful treatment of cancer. The identification of tumor-coded genes and how they contribute to the progression of cancer is required to improve patient outcomes. Recently, cells that have undergone the epithelial-mesenchymal transition (EMT), which share characteristics with cancer stem cells (CSC) have been implicated to play a role in drug resistance and metastasis of several types of cancer. In this review, we discuss the relationship among transglutaminase 2 (TG2), the EMT, and CSCs in inflammation and cancer. TG2 is a structurally and functionally complex protein implicated in such diverse processes as tissue fibrosis, wound healing, apoptosis, neurodegenerative disorders, celiac disease, atherosclerosis and cancer. Depending on the cellular context, TG2 can either promote or inhibit cell death. Increased expression of TG2 in several types of cancer cells has been associated with increased cell invasiveness, cell survival and decreased survival of patients with cancer. Down-regulation of TG2 by small interfering RNA (siRNA) or its inhibition by small molecule inhibitors has been shown to significantly enhances the therapeutic efficacy of anticancer drugs and inhibit metastatic spread. In addition, TG2-regulated pathways are involved in promoting or protecting normal and tumor cells from death-induced signaling. We discuss the contribution of TG2-regulated pathways to the development of drug resistance and progression to metastatic disease and the therapeutic potential of TG2 for treating advanced-stage cancer.


Subject(s)
GTP-Binding Proteins/physiology , Inflammation/enzymology , Neoplasms/enzymology , Transglutaminases/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , GTP-Binding Proteins/genetics , Genetic Therapy , Humans , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Protein Glutamine gamma Glutamyltransferase 2 , RNA, Small Interfering/therapeutic use , Transglutaminases/genetics
12.
Mol Cancer Res ; 7(10): 1605-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19808905

ABSTRACT

Integrin alpha6beta4 is linked to cancer cell motility and invasion in aggressive and metastatic cancer cells. In this study, we showed that expression of the beta4 integrin in MDA-MB-435 cancer cells (MDA-MB-435/beta4) leads to a dramatic increase in expression of a metastasis-promoting factor, S100A4, as determined by affymetrix gene chip microarray, quantitative real-time PCR, and Western blot analysis. Alternatively, knocking down beta4 integrin expression in MDA-MB-231 breast carcinoma cells by shRNA reduced the level of S100A4 expression. The mechanism by which alpha6beta4 enhances S100A4 expression involves Src, Akt, and NFAT. We have further shown that Y1494, a tyrosine residue of the ITIM motif in the cytoplasmic domain of the beta4 integrin subunit, is essential for alpha6beta4-dependent S100A4 expression. Reduction of S100A4 expression by shRNA blocked migration, invasion, and anchorage-independent growth of MDA-MB-435/beta4, SUM-159, and MDA-MB-231 cells. These studies define a novel mechanism by which integrin alpha6beta4 promotes cancer cell motility and invasion, and provides insight into how S100A4 expression is regulated in cancer cells.


Subject(s)
Integrin alpha6beta4/genetics , Neoplasm Invasiveness/genetics , Neoplasms/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , S100 Proteins/genetics , Signal Transduction/genetics , Amino Acid Motifs/physiology , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Integrin alpha6beta4/chemistry , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasms/genetics , Neoplasms/physiopathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism , Up-Regulation/genetics , src-Family Kinases
13.
Cancer Prev Res (Phila) ; 1(5): 385-91, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19138983

ABSTRACT

Curcumin, a polyphenol natural product isolated from the rhizome of the plant Curcuma longa, has emerged as a promising anticancer therapeutic agent. However, the mechanism by which curcumin inhibits cancer cell functions such as cell growth, survival, and cell motility is largely unknown. We explored whether curcumin affects the function of integrin alpha(6)beta(4), a laminin adhesion receptor with an established role in invasion and migration of cancer cells. Here we show that curcumin significantly reduced alpha(6)beta(4)-dependent breast cancer cell motility and invasion in a concentration-dependent manner without affecting apoptosis in MDA-MB-435/beta4 (beta(4)-integrin transfectants) and MDA-MB-231 breast cancer cell lines. Further, curcumin selectively reduced the basal phosphorylation of beta(4) integrin (Y1494), which has been reported to be essential in mediating alpha(6)beta(4)-dependent phosphatidylinositol 3-kinase activation and cell motility. Consistent with this finding, curcumin also blocked alpha(6)beta(4)-dependent Akt activation and expression of the cell motility-promoting factor ENPP2 in MDA-MB-435/beta4 cell line. A multimodality approach using curcumin in combination with other pharmacologic inhibitors of alpha(6)beta(4) signaling pathways showed an additive effect to block breast cancer cell motility and invasion. Taken together, these findings show that curcumin inhibits breast cancer cell motility and invasion by directly inhibiting the function of alpha(6)beta(4) integrin, and suggest that curcumin can serve as an effective therapeutic agent in tumors that overexpress alpha(6)beta(4).


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Cell Movement/drug effects , Curcumin/pharmacology , Integrin alpha6beta4/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Carcinoma/metabolism , Carcinoma/prevention & control , Cell Adhesion/drug effects , Cell Movement/genetics , Curcumin/administration & dosage , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Humans , Indoles/administration & dosage , Indoles/pharmacology , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Integrin alpha6beta4/physiology , Neoplasm Invasiveness , Oncogene Protein v-akt/antagonists & inhibitors , Phosphorylation/drug effects , Piperazines/administration & dosage , Piperazines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Tumor Cells, Cultured
14.
Brain Res ; 1148: 53-61, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17359944

ABSTRACT

Celecoxib is a selective cyclooxygenase-2 (COX-2) inhibitor used in the treatment of osteoarthritis and rheumatoid arthritis with fewer gastrointestinal toxicities compared to traditional non-steroidal anti-inflammatory drugs. Voltage-gated Na(+) channels in primary sensory neurons play an important role in the pathogenesis of various pain conditions. We examined the effects of celecoxib on tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) Na(+) currents in acutely dissociated rat dorsal root ganglion neurons. Celecoxib suppressed both currents in dose- and frequency-dependent manner. The apparent dissociation constants (K(d)) for TTX-S and TTX-R Na(+) currents measured at 0 mV from a holding potential of -80 mV were estimated to be 5.6 and 19.5 microM, respectively. Celecoxib slightly slowed inactivation kinetics of TTX-S Na(+) current, but made it much faster in TTX-R Na(+) current. Celecoxib shifted the activation voltage of TTX-S Na(+) current to a depolarizing direction, but not that of TTX-R Na(+) current. Celecoxib caused a hyperpolarizing shift of the steady-state inactivation curve in both Na(+) currents to a great extent. In addition celecoxib reduced the maximal availability of both Na(+) channels. Thus celecoxib appears to bind to both inactivated and resting Na(+) channels. Celecoxib slowed the recovery of both Na(+) channels from inactivation. All these effects combined would suppress the excitability of sensory neurons. Thus, beside COX-2 inhibition, the Na(+) channel inhibition is considered to contribute to celecoxib analgesia.


Subject(s)
Ganglia, Spinal/drug effects , Neurons, Afferent/drug effects , Nociceptors/drug effects , Pain/drug therapy , Pyrazoles/pharmacology , Sodium Channels/drug effects , Sulfonamides/pharmacology , Animals , Animals, Newborn , Celecoxib , Cells, Cultured , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Ganglia, Spinal/metabolism , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons, Afferent/metabolism , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Tetrodotoxin/pharmacology
15.
Brain Res ; 1072(1): 62-71, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16423329

ABSTRACT

Nordihydroguaiaretic acid (NDGA) is a lipoxygenase (LO) inhibitor with a strong antioxidant activity. It attenuates nociceptive responses produced by various stimuli, which has been ascribed to its LO inhibition. Primary sensory neurons express multiple Na+ channels that are important in processing normal and abnormal nociception. We examined the effects of NDGA on tetrodotoxin-sensitive and tetrodotoxin-resistant Na+ currents in rat dorsal root ganglion neurons. NDGA inhibited both types of Na+ currents concentration dependently and reversibly. Both activation and inactivation time courses were slowed by NDGA, which were not reversible. NDGA produced a hyperpolarizing shift of the steady-state inactivation curves and reduced the maximal availability of both Na+ currents, indicating that it blocks both inactivated and resting Na+ channels. NDGA shifted the conductance-voltage curves of both Na+ currents toward a depolarizing direction and increased the slope factors of the curves. The recovery of Na+ channels from inactivation was retarded by NDGA. All these effects will reduce the excitability of sensory neurons and should be taken into account when it comes to the antinociceptive effects of NDGA.


Subject(s)
Ganglia, Spinal/physiology , Masoprocol/pharmacology , Neurons/physiology , Sodium Channels/physiology , Animals , Cells, Cultured , Lipoxygenase Inhibitors/pharmacology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Sodium Channels/drug effects , Tetrodotoxin/pharmacology
16.
Brain Res ; 1062(1-2): 39-47, 2005 Nov 16.
Article in English | MEDLINE | ID: mdl-16256960

ABSTRACT

Anandamide, the ethanolamide of arachidonic acid, is an endogenous cannabinoid. It is an agonist at CB1 and CB2 cannabinoid receptors as well as the vanilloid receptor, VR1. It is analgesic in inflammatory and neuropathic pain. Both central and peripheral mechanisms are considered to participate in its analgesia. Primary sensory neurons express Na+ currents that are involved in the pathogenesis of pain. We examined the effect of anandamide on tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) Na+ currents in rat dorsal root ganglion neurons. Anandamide inhibited both Na+ currents in a concentration-dependent manner. At a membrane potential of -80 mV, the current inhibition was greater in TTX-S than TTX-R currents (K(d); 5.4 microM vs. 38.4 microM). The activation and inactivation became faster in TTX-R current but not in TTX-S current. Anandamide did not alter the activation voltage in either type of current. It, however, produced a hyperpolarizing shift of the steady-state inactivation voltage in both types of currents. The maximum availability at a large negative potential was not reduced by anandamide. Thus, anandamide seems to affect inactivated Na+ channels rather than resting channels. The inhibition of Na+ currents was not reversed by AM 251 (a CB1 antagonist), AM 630 (a CB2 antagonist) or capsazepine (a VR1 antagonist), suggestive of a direct action of anandamide on Na+ channels. The inhibition of Na+ currents in sensory neurons may contribute to the anandamide analgesia.


Subject(s)
Analgesics/pharmacology , Arachidonic Acids/pharmacology , Ganglia, Spinal/drug effects , Neurons, Afferent/drug effects , Sodium Channels/drug effects , Animals , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids , Ganglia, Spinal/cytology , In Vitro Techniques , Ion Channel Gating/drug effects , Patch-Clamp Techniques , Polyunsaturated Alkamides , Rats , Sodium Channel Blockers/pharmacology , Sodium Channels/classification , Tetrodotoxin/pharmacology
17.
Brain Res ; 1045(1-2): 134-41, 2005 May 31.
Article in English | MEDLINE | ID: mdl-15910771

ABSTRACT

Resveratrol, a phytoalexin found in grapevines, exerts neuroprotective, cancer chemopreventive, antiinflammatory and cardioprotective activities. Studies have also shown that resveratrol exhibits analgesic effects. Cyclooxygenase inhibition and K+ channel opening have been suggested as underlying mechanisms for the resveratrol-induced analgesia. Here, we investigated the effects of resveratrol on tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) Na+ currents in rat dorsal root ganglion (DRG) neurons. Resveratrol suppressed both Na+ currents evoked at 0 mV from -80 mV. TTX-S Na+ current (K(d), 72 microM) was more susceptible to resveratrol than TTX-R Na+ current (K(d), 211 microM). Although the activation voltage of TTX-S Na+ current was shifted in the depolarizing direction by resveratrol, that of TTX-R Na+ current was not. Resveratrol caused a hyperpolarizing shift of the steady-state inactivation voltage and slowed the recovery from inactivation of both Na+ currents. However, no frequency-dependent inhibition of resveratrol on either type of Na+ current was observed. The suppression and the unfavorable effects on the kinetics of Na+ currents in terms of the excitability of DRG neurons may make a great contribution to the analgesia by resveratrol.


Subject(s)
Ganglia, Spinal/drug effects , Neurons, Afferent/drug effects , Nociceptors/drug effects , Pain/drug therapy , Sodium Channels/drug effects , Stilbenes/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ganglia, Spinal/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , NAV1.7 Voltage-Gated Sodium Channel , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons, Afferent/physiology , Neuropeptides/drug effects , Neuropeptides/metabolism , Nociceptors/physiology , Pain/metabolism , Pain/physiopathology , Rats , Resveratrol , Sodium Channels/metabolism
18.
Brain Res ; 1008(1): 81-91, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15081385

ABSTRACT

Free fatty acids (FFAs), especially polyunsaturated fatty acids (PUFAs), are potent modulators of muscle-type sodium channels. It is not known if they also modulate sodium channels of sensory neurons. In this study, we investigated the effects of FFAs on the fast tetrodotoxin-sensitive (fTTX-S) and the slow tetrodotoxin-resistant (sTTX-R) sodium currents in rat dorsal root ganglion neurons. At a holding potential of -80 mV, PUFAs potently inhibited fTTX-S current, but monounsaturated fatty acids (MUFAs) and saturated fatty acids (SFAs) to a lesser extent. All FFAs initially increased sTTX-R current, and then decreased it slightly. PUFAs and MUFAs produced a hyperpolarizing shift of the steady-state inactivation voltage for both types of sodium currents. The shift generally increased with the number of unsaturated bonds. FFAs did not change the maximum amplitude of fTTX-S current, but increased that of sTTX-R current. Most FFAs shifted the activation voltage for fTTX-S current in the hyperpolarizing direction, which was not dependent on the degree of unsaturation. MUFAs and SFAs shifted the activation voltage for sTTX-R current in the hyperpolarizing direction, but PUFAs were without effect. The modulation of sodium currents by FFAs, especially PUFAs, may have considerable impact on the excitability of sensory neurons.


Subject(s)
Fatty Acids/pharmacology , Ganglia, Spinal/cytology , Neurons/drug effects , Sodium Channels/physiology , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Electric Conductivity , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Neurons/classification , Neurons/physiology , Patch-Clamp Techniques/methods , Rats , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Tetrodotoxin/pharmacology
19.
Brain Res ; 1006(2): 168-76, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15051520

ABSTRACT

The effects of various nucleotides on the fast tetrodotoxin-sensitive (f-TTX-S) and the slow tetrodotoxin-resistant (s-TTX-R) sodium currents in rat dorsal root ganglion (DRG) neurons were investigated using the patch-clamp technique. Nucleoside triphosphates (NTPs; ATP, GTP, UTP and CTP) and nucleoside diphosphates (NDPs; ADP, GDP, UDP and CDP) decreased f-TTX-S current, whereas they increased s-TTX-R current, when currents were evoked by step depolarizations to 0 mV from a holding potential of -80 mV. NTPs and NDPs shifted both the conductance-voltage relationship curve and the steady-state inactivation curve in the hyperpolarizing direction in both types of sodium currents. Most of them also increased the maximum conductance of s-TTX-R current. ITP, a derivative of ribonucleotide, and dTTP, a deoxyribonucleotide, modulated both types of sodium currents similarly to NTPs and NDPs. However, nucleoside monophosphates (NMPs; AMP, GMP, UMP and CMP) and adenosine had little or no effect on either type of sodium current. Therefore, it seems that nucleotides, regardless of the kind of base, should have two or more phosphates to be able to modulate sodium currents in DRG neurons. Extracellular nucleotides with di- or tri-phosphates would influence the perception by modulating sodium currents in sensory neurons. Particularly, the increase of the maximum conductance and the hyperpolarizing shift of the conductance-voltage relationship of s-TTX-R sodium current would result in an intensified nociception.


Subject(s)
Ganglia, Spinal/cytology , Neurons, Afferent/drug effects , Nucleotides/pharmacology , Sodium Channels/drug effects , Sodium/metabolism , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Electric Conductivity , Electric Stimulation/methods , Evoked Potentials/drug effects , Evoked Potentials/radiation effects , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Neurons, Afferent/physiology , Patch-Clamp Techniques/methods , Rats , Sodium Channels/classification , Sodium Channels/physiology , Sodium Channels/radiation effects , Tetrodotoxin/pharmacology
20.
Brain Res ; 992(1): 120-7, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14604780

ABSTRACT

The effects of diclofenac, a nonsteroidal anti-inflammatory drug (NSAID), on the fast tetrodotoxin-sensitive (TTX-S) and the slow tetrodotoxin-resistant (TTX-R) sodium currents in rat dorsal root ganglion neurons were investigated using the whole-cell patch-clamp method. Diclofenac suppressed both sodium currents in a dose-dependent manner. The apparent dissociation constants for the diclofenac suppression of TTX-S and TTX-R sodium currents were estimated to be 14 and 97 microM, respectively, at a holding potential of -80 mV. Diclofenac had no effect on the kinetic parameters of the activation process in either type of sodium current. However, diclofenac produced shifts of the steady-state inactivation curves in the hyperpolarizing direction in both types of sodium currents in a dose-dependent manner. At sufficiently negative holding potentials, the inhibitory effects of diclofenac on both types of sodium currents were minimal. The results suggested that diclofenac might bind to sodium channels with a greater affinity when they are in the inactivated state than when they are in the resting state. Effects of other NSAIDs (acetylsalicylic acid, antipyrin, indomethacin and flufenamic acid) on sodium currents were tested. Among these, only flufenamic acid suppressed the sodium currents to a considerable extent. Thus, the chemical structure of each NSAID, not the inhibition of cyclooxygenase, seems to be an important determinant in the sodium current inhibition. The suppression of sodium currents in sensory neurons by diclofenac and flufenamic acid would contribute to their analgesic activity in addition to their inhibition of cyclooxygenase.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Membrane Potentials/drug effects , Neurons/drug effects , Sodium/metabolism , Animals , Animals, Newborn , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Membrane Potentials/physiology , Neurons/physiology , Patch-Clamp Techniques , Rats , Sodium Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...