Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Apoptosis ; 26(3-4): 184-194, 2021 04.
Article in English | MEDLINE | ID: mdl-33515314

ABSTRACT

Previously we have shown inhibition of endometrial cancer cell growth with progesterone and calcitriol. However, the mechanisms by which the two agents attenuate proliferation have not been well characterized yet. Herein, we investigated how progesterone and calcitriol induce apoptosis in cancer cells. DNA fragmentation was upregulated by progesterone and calcitriol in ovarian and endometrial cancer cells. Time-dependent treatment of ovarian cancer cells, ES-2, and TOV-21G with progesterone enhanced caspase -8 activity after 12 h, whereas OV-90, TOV-112D, HEC-1A, and HEC-59 cells showed increased activity after 24 h. Caspase 9 activity was increased in all cell lines after 24 h treatment with calcitriol. Pretreatment of cancer cells with a caspase-8 inhibitor (z-IETD-fmk) or caspase-9 inhibitor (Z-LEHD-fmk) significantly attenuated progesterone and calcitriol induced caspase-8 and caspase-9 expression, respectively. The expression of FasL, Fas, FAD, and pro-caspase-8, which constitute the death-inducing signaling complex (DISC), was upregulated in progesterone treated cancer cells. Knockdown of FAS or FADD with specific siRNAs significantly blocked progesterone-induced caspase-8. Cleavage of the BID was not affected by caspase-8 activation suggesting the absence of cross-talk between caspase-8 and caspase-9 pathways. Calcitriol treatment decreased mitochondrial membrane potential and increased the release of cancer cytochrome C. These findings indicate that progesterone induces apoptosis through activation of caspase-8 and calcitriol through caspase-9 activation in cancer cells. A combination of progesterone-calcitriol activates both extrinsic and intrinsic apoptotic pathways in cancer cells.


Subject(s)
Apoptosis/drug effects , Caspases , Endometrial Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Progesterone/pharmacology , Calcitriol/metabolism , Caspase 8/drug effects , Caspase 8/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/drug effects , Cytochromes c/metabolism , Death Domain Receptor Signaling Adaptor Proteins/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Domain Superfamily/drug effects , Endometrial Neoplasms/drug therapy , Fas Ligand Protein/drug effects , Fas Ligand Protein/metabolism , Female , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Ovarian Neoplasms/drug therapy , Signal Transduction/drug effects , fas Receptor/drug effects , fas Receptor/metabolism
2.
Biomedicines ; 8(4)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32244545

ABSTRACT

: Initially, patients that respond to cisplatin (DDP) treatment later relapse and develop chemoresistance. Agents that enhance DDP effectiveness will have a significant impact on cancer treatment. We have shown pronounced inhibitory effects of the progesterone-calcitriol combination on endometrial and ovarian cancer cell growth. Here, we examined whether and how progesterone-calcitriol combination potentiates DDP anti-tumor effects in cancer cells. Ovarian and endometrial cancer cells treated with various concentrations of DDP showed a concentration-dependent decrease in cell proliferation. Concurrent treatment of cells with DDP and progesterone-calcitriol ombination potentiated anticancer effects of DDP compared to DDP-calcitriol, or DDP-progesterone treated groups. The anticancer effects were mediated by increased caspase-3, BAX, and decreased BCL2 and PARP-1 expression in DDP and progesterone-calcitriol combination-treated cells. Stimulation of the PI3K/AKT and MAPK/ERK pathways seen in cancer cells was reduced in DDP-progesterone-calcitriol treated cells. Pretreatment of cells with specific inhibitors further diminished AKT and ERK expression. Furthermore, progesterone-calcitriol potentiated the anti-growth effects of DDP on cancer cells by attenuating the expression of SMAD2/3, multidrug resistance protein- 1 (MDR-1), and ABC transporters (ABCG1, and ABCG2), thereby impeding the efflux of chemo drugs from cancer cells. These results suggest a potential clinical benefit of progesterone-calcitriol combination therapy when used in combination with DDP.

3.
Molecules ; 24(20)2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31623109

ABSTRACT

Nitric oxide (NO) is implicated in several biological processes, including cancer progression. At low concentrations, it promotes cell survival and tumor progression, and at high concentrations it causes apoptosis and cell death. Until now, the impact of NO donors has not been investigated on human endometrial tumors. Four cancer cell lines were exposed to different concentrations of DETA/NO for 24 to 120 h. The effects of DETA/NO on cell proliferation and invasion were determined utilizing MTS and Boyden chamber assays, respectively. The DETA/NO induced a dose and time-dependent reduction in cell viability by the activation of caspase-3 and cell cycle arrest at the G0/G1 phase that was associated with the attenuated expression of cyclin-D1 and D3. Furthermore, the reduction in the amount of CD133-expressing cancer stem-like cell subpopulation was observed following DETA/NO treatment of cells, which was associated with a decreased expression of stem cell markers and attenuation of cell invasiveness. To understand the mechanisms by which DETA/NO elicits anti-cancer effects, RNA sequencing (RNA-seq) was used to ascertain alterations in the transcriptomes of human endometrial cancer cells. RNA-seq analysis revealed that 14 of the top 21 differentially expressed genes were upregulated and seven were downregulated in endometrial cancer cells with DETA/NO. The genes that were upregulated in all four cell lines with DETA/NO were the tumor suppressors Ras association domain family 1 isoform A (RASSF1) and Cyclin-dependent kinase inhibitor 1A (CDKN1A). The expression patterns of these genes were confirmed by Western blotting. Taken together, the results provide the first evidence in support of the anti-cancer effects of DETA/NO in endometrial cancer.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation/drug effects , Nitric Oxide Donors/pharmacology , Tumor Suppressor Proteins/genetics , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Knockdown Techniques , Humans , Nitric Oxide/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism
4.
Transl Cancer Res ; 8(4): 1423-1437, 2019 Aug.
Article in English | MEDLINE | ID: mdl-35116885

ABSTRACT

BACKGROUND: Strategies are needed to coordinately block drivers and induce suppressors of cancer to reduce incidence and improve outcomes for individuals with inherited or acquired risk. We previously reported the chemopreventive and therapeutic efficacy of the combination of progestin and calcitriol in transformed and malignant endometrioid endometrial cancer (EC) and in ovarian cancer models involving attenuated expression of TGF-ß signaling proteins and progestin-mediated inhibition of calcitriol-induced CYP24A1 expression. This study aims to expand the applications for this combination to other subtypes of endometrial and ovarian cancers, including those with mutations in ARID1A or PIK3CA, DNA mismatch repair (MMR) deficiency or BRCA1 null status. METHODS: Ovarian and EC cell lines of different histotypes were cultured with either progesterone, calcitriol, or the combination of progesterone and calcitriol for 3 or 5 days. The end points for this in vitro investigation included assessments of cell growth by (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assays and the expression of TGF-ß ligands, receptors, SMAD proteins and CYP24A1 by western blotting. RESULTS: Treatment of ovarian clear cell carcinoma, endometrioid carcinoma, papillary serous adenocarcinoma, BRCA1 null, and DNA MMR deficient EC cell lines with progesterone alone or in combination with calcitriol inhibited cell growth and expression of TGF-ß1, TGF-ß2, TGF-Rß1, TGF-ßR2, pSMAD2/3 and CYP24A1. Expression of TGF-ßR3, SMAD-4, progesterone receptor (PR) and vitamin-D receptor (VDR) was not altered in any cell line tested except, ES-2, where VDR expression was upregulated in response to treatment. CONCLUSIONS: These results suggest that progesterone alone and progesterone-calcitriol combination have broad application in both chemopreventive and therapeutic settings that merit further development in a wide variety of ovarian and ECs, including those derived from germline or somatic mechanisms. Moreover, our data suggest that TGF-ß signaling proteins and CYP24A1 may be effective surrogate markers indicative of treatment response.

5.
Int J Gynecol Pathol ; 38(5): 449-458, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30028355

ABSTRACT

TP53 gene mutations are known to manifest in distinct p53 immunohistochemical staining patterns; overexpression, wild-type, and null. These stratified staining patterns are routinely utilized in subtyping ovarian cancer subtypes. Three ovarian cancer cell lines were used in the construction of an immunohistochemical p53 expression pattern control panel that highlight respective TP53 mutation status. The cell line control panel sections demonstrated consistent clean and easily interpretable p53 immunohistochemical staining. Procured resection, biopsy, and cytologic specimens were submitted along with either standard control tissue or a p53 cell line control panel to pathologists of varying experience for interrater reliability analysis. Individual interrater reliability was near-perfect and was improved with the p53 cell line control panel when compared with the tissue control. The cell line control panel demonstrated decreased misinterpretation of null expression pattern as wild-type. Next-generation sequencing analysis was performed on the cell lines and select cases, in which there was discordance in p53 expression pattern interpretation. Next-generation sequencing analysis demonstrated low-frequency variant mutations in some cases in which there was reviewer discordance. This study suggests the addition of a p53 cell line expression pattern control panel could potentially increase p53 interpretation accuracy for ovarian cancer subtypes. We developed a cell line-based p53 control panel that has the potential to increase individual interrater reliability for p53 immunohistochemical expression pattern determination, support immunohistochemical optimization, and direct submission of difficult to interpret p53 staining cases to next-generation sequencing.


Subject(s)
Ovarian Neoplasms/chemistry , Tumor Suppressor Protein p53/analysis , Cell Line, Tumor , Female , Genes, p53 , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Mutation
6.
Exp Cell Res ; 370(2): 663-670, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30036538

ABSTRACT

The matrix metalloproteinases (MMPs) are implicated in tumor invasion and metastasis. Given their multiple tumor promoting roles, MMPs are promising targets for the treatment of metastatic cancer. Using a siRNA library screen of 140 membrane trafficking genes, we identified 41 genes in HEC-1B and 36 in Ishikawa cancer cells that decreased metalloproteinases activity. The 16 genes common in both cancer cell lines that decreased MMPs activity are involved in cargo sorting, vesicle formation and vesicle recycling. The top two genes clathrin-B and cofilin-1 were chosen for post hoc functional studies. Higher expression of both genes was confirmed in cancer cells and knockdown with respective siRNAs inhibited their invasive potential and matrix metalloproteinases activity. Membrane Type 1- Matrix Metalloproteinase (MT1-MMP) is a master switch proteinase and regulator of invasion and metastasis. A marked decrease in MT1-MMP expression and activity was seen in clathrin-B and cofilin-1 knockdown cancer cells which was associated with a marked decreased expression of invadopodia formation proteins. Our results suggest that the decreased expression of clathrin-B and cofilin-1 decreases the expression of MT1-MMP and results in attenuation of MT1-MMP at the cell surface, thus inhibiting tumor cell invasion and metastasis.


Subject(s)
Cofilin 1/genetics , Endometrial Neoplasms/genetics , RNA Interference/physiology , Sesquiterpenes/metabolism , Early Detection of Cancer/methods , Endometrial Neoplasms/diagnosis , Extracellular Matrix/metabolism , Female , Humans , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases/metabolism , Metalloendopeptidases , Neoplasm Invasiveness/genetics
7.
J Cell Biochem ; 119(9): 7515-7524, 2018 09.
Article in English | MEDLINE | ID: mdl-29797601

ABSTRACT

Hypoxia-inducible factor-1alpha (HIF-1α) is aberrantly upregulated in tumors and implicated in angiogenesis, metastasis, and drug resistance. Therefore, developing treatments that target HIF-1α may be a viable therapeutic approach. In Traditional Chinese Medicine (TCM), Scutellaria baicalensis (SB) is used for the treatment of cancer but the anti-cancer mechanisms are not known. We examined the effects of SB on HIF-1α expression in ovarian cancer (OC) cell lines grown under normoxic and hypoxic conditions. SB treatment attenuated HIF-1α expression in cancer cell lines. Treatment of cells with cycloheximide (CHX) reduced HIF-1α levels similar to cells treated with SB. Furthermore, SB-induced HIF-1α inhibition was abrogated by the proteasomal inhibitor MG132 and a lysosome inhibitor, chloroquine. Activation of PI3K/AKT and MAPK/ERK seen in OC cells was reduced with SB. Pretreatment of cells with LY294002 (phosphoinositide 3-kinase inhibitor) and PD98059 (mitogen-activated protein kinase inhibitor) reduced HIF-1α expression comparable to SB-treated cells. SB potentiated the anti-growth effects of cisplatin on OC cells by attenuating the expression of HIF-1α, ABCG1, and ABCG2. Taken together, the findings suggest that targeting HIF-1α with SB could be an effective treatment strategy for cancer and SB can improve the sensitivity of cancer cells to cisplatin, which is a major challenge in therapy for ovarian tumors.


Subject(s)
Cisplatin/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ovarian Neoplasms/drug therapy , Plant Extracts/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Scutellaria baicalensis , Signal Transduction , Treatment Outcome
8.
Oncotarget ; 8(69): 113583-113597, 2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29371931

ABSTRACT

Previously, we have demonstrated that progesterone and calcitriol synergistically inhibit growth of endometrial and ovarian cancer by enhancing apoptosis and causing cell cycle arrest. Metastasis is the main reason of mortality in cancer patients. Activation of ADP-Ribosylation Factor 6 (ARF6), Neural Precursor cell expressed Developmentally Downregulated 9 (NEDD9), and Membrane-Type-1 Matrix Metalloproteinase (MT1-MMP) have been implicated in promoting tumor growth and metastasis. We examined the effects of progesterone, calcitriol and progesterone-calcitriol combination on metastasis promoting proteins in endometrial cancer. Expression of ARF6, NEDD9, and MT1-MMP was enhanced in advanced-stage endometrial tumors and in cancer cell lines compared to normal tissues and immortalized EM-E6/E7-TERT endometrial epithelial cells. Knockdown of these proteins significantly inhibited the invasiveness of the cancer cells. The expression levels of all three proteins was reduced with progesterone and progesterone-calcitriol combination treatment, whereas calcitriol alone showed no effect on their expression but moderately decreased MT1-MMP activity. Fluorescence microscopy showed membrane expression of MT1-MMP in vehicle and calcitriol-treated endometrial cancer cells. However, progesterone and calcitriol-progesterone combination treatment revealed MT1-MMP in the cytoplasm. Furthermore, progesterone and calcitriol reduced the activity of MT1-MMP, MMP-9, and MMP-2. In addition, invadopodia regulatory proteins were attenuated in both progesterone and progesterone-calcitriol combination treated cells as well as in MT1-MMP knockdown cells. Thus, targeting the aberrant MT1-MMP signaling with progesterone-calcitriol may be a novel approach to impede MT1-MMP mediated cancer dissemination and may have therapeutic benefits for endometrial cancer patients.

9.
Oncotarget ; 7(47): 77576-77590, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27769055

ABSTRACT

Here, we evaluated the expression of CYP24A1, a protein that inactivates vitamin D in tissues. CYP24A1 expression was increased in advanced-stage endometrial tumors compared to normal tissues. Similarly, endometrial cancer cells expressed higher levels of CYP24A1 than immortalized endometrial epithelial cells. RT-PCR and Western blotting were used to examine CYP24A1 mRNA and protein levels in endometrial cancer cells after 8, 24, 72, and 120 h of exposure to progesterone, progestin derivatives and calcitriol, either alone or in combination. Progestins inhibited calcitriol-induced expression of CYP24A1 and splice variant CYP24SV mRNA and protein in cancer cells. Furthermore, actinomycin D, but not cycloheximide, blocked calcitriol-induced CYP24A1 splicing. siRNA-induced knockdown of CYP24A1 expression sensitized endometrial cancer cells to calcitriol-induced growth inhibition. These data suggest that CYP24A1 overexpression reduces the antitumor effects of calcitriol in cancer cells and that progestins may be beneficial for maintaining calcitriol's anti-endometrial cancer activity.


Subject(s)
Calcitriol/pharmacology , Endometrial Neoplasms/pathology , Progesterone/pharmacology , Vitamin D3 24-Hydroxylase/genetics , Vitamin D3 24-Hydroxylase/metabolism , Adult , Aged , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Middle Aged , Neoplasm Grading
10.
Oncotarget ; 7(43): 69733-69748, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27626172

ABSTRACT

Nestin, an intermediate filament protein and a stem cell marker is expressed in several tumors. Until recently, little was known about the expression levels and the role of Nestin in endometrial cancer. Compared to the immortalized endometrial epithelial cell line EM-E6/E7-TERT, endometrial cancer cell lines express high to moderate levels of Nestin. Furthermore, endometrial tumors and tumor cell lines have a cancer stem-like cell subpopulation expressing CD133. Among the cancer lines, AN3CA and KLE cells exhibited both a significantly higher number of CD133+ cells and expressed Nestin at higher levels than Ishikawa cells. Knockdown of Nestin in AN3CA and KLE increased cells in G0/G1 phase of the cell cycle, whereas overexpression in Ishikawa decreased cells in G0/G1 phase and increased cells in S-phase. Nestin knockdown cells showed increased p21, p27, and PNCA levels and decreased expression of cyclin-D1 and D3. In contrast, Nestin overexpression revealed an inverse expression pattern of cell cycle regulatory proteins. Nestin knockdown inhibited cancer cell growth and invasive potential by downregulating TGF-ß signaling components, MMP-2, MMP-9, vimentin, SNAIL, SLUG, Twist, N-cadherin, and upregulating the epithelial cell marker E-cadherin whereas the opposite was observed with Nestin overexpressing Ishikawa cells. Nestin knockdown also inhibited, while overexpression promoted invadopodia formation and pFAK expression. Knockdown of Nestin significantly reduced tumor volume in vivo. Finally, progesterone inhibited Nestin expression in endometrial cancer cells. These results suggest that Nestin can be a therapeutic target for cancer treatment.


Subject(s)
Endometrial Neoplasms/pathology , Nestin/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Cadherins/analysis , Cell Line, Tumor , Down-Regulation , Epithelial-Mesenchymal Transition , Female , G1 Phase Cell Cycle Checkpoints , Humans , Neoplasm Invasiveness , Progesterone/pharmacology
11.
Acta Pol Pharm ; 73(3): 565-78, 2016.
Article in English | MEDLINE | ID: mdl-27476273

ABSTRACT

Hispidulin is well-known natural bioactive flavone on behalf of its pharmacological aspects. This review contains data on isolation, synthetic methodology, pharmacokinetics and bioactivities of hispidulin. The article provides a critical assessment of present knowledge about hispidulin with some clear conclusions, perspectives and directions for future research in potential applications.


Subject(s)
Flavones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Flavones/chemical synthesis , Flavones/pharmacokinetics , Humans , Hypnotics and Sedatives/antagonists & inhibitors , Mitochondria , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Protective Agents/pharmacokinetics , Protective Agents/pharmacology
12.
Gynecol Oncol ; 143(1): 159-167, 2016 10.
Article in English | MEDLINE | ID: mdl-27106018

ABSTRACT

OBJECTIVES: Previously we have shown in endometrial cells that progesterone (P4) and calcitriol (CAL, 1,25(OH)2D3) synergistically promote apoptosis and that progestins induce expression of the vitamin D receptor. In the current study we examined the progestin/vitamin D combination in ovarian cells and searched for other progestin-related effects on vitamin D metabolism that may underlie the novel interaction between progestins and vitamin D, including whether progestins inhibit CYP24A1, the enzyme that renders CAL inactive. METHODS: We investigated the impact of P4 on CAL-induced CYP24A1 expression in cancer cell lines expressing progesterone receptors (PRs), [OVCAR-5, OVCAR-3-PGR (PR-transfected OVCAR-3 ovarian line), and T47D-WT, T47D-A and T47D-B (breast lines expressing PRs or individual PR isoforms)] or lines that do not express PRs (OVCAR-3 and T47D-Y). We examined CYP24A1 expression using RT-PCR and western blotting, and apoptosis by TUNEL. We also investigated P4 inhibition of Cyp24a1 in ovaries from CAL-treated mice. RESULTS: CAL treatment induced CYP24A1 expression. When co-treated with P4, cell lines expressing PRs showed marked inhibition of CYP24A1 expression (p<0.001), along with increased apoptosis (p<0.01); cells not expressing PRs did not. Mouse ovaries showed a significant reduction in CAL-induced Cyp24a1 mRNA (p<0.001) and protein (p<0.01) in response to P4. CONCLUSIONS: We show for the first time that progestins and vitamin D synergistically reduce cell viability and induce apoptosis in ovarian cells and that progestins PR-dependently inhibit CAL-induced CYP24A1, thus extending CAL activity. The combination of progestins and vitamin D deserves further consideration as a strategy for inhibiting ovarian carcinogenesis.


Subject(s)
Calcitriol/pharmacology , Chemoprevention , Ovarian Neoplasms/drug therapy , Progesterone/pharmacology , Vitamin D3 24-Hydroxylase/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Female , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/pathology , Ovary/enzymology , Receptors, Progesterone/analysis , Receptors, Progesterone/physiology
13.
Gynecol Oncol ; 140(3): 503-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718725

ABSTRACT

OBJECTIVE: Evidence of potential prognostic and predictive value for nestin was investigated in well-annotated uterine cancers (UCs). METHODS: Nestin expression and previously-published biomarkers were evaluated by immunohistochemistry (IHC) in UC tissue microarrays. Biomarkers were categorized as low vs. high, and nestin was cut at 10% positive staining. Relationship between nestin and clinicopathologic factors, biomarkers and outcome were evaluated using exact/log-rank testing or logistic/Cox modeling. RESULTS: There were 323 eligible cases, 34% had advanced stage disease, 37% had type II disease, and 5% were carcinosarcomas. High nestin, observed in 19% of cases, was more common in advanced vs. early stage disease, type II cancers or uterine carcinosarcoma vs. type I cancers, grade 3 disease, positive lymphovascular space invasion (LVSI) and tumors >6cm (p<0.05). Nestin was inversely correlated with ER, PR and TFF3, and correlated with p53 and IMP3. Women with high vs. low nestin had worse progression-free survival (PFS) and cancer-specific survival overall, and worse PFS in the subset who received no adjuvant therapy or radiation, or had early stage, type I disease or tumors with both low and high ER, PR, TFF3, PTEN, p53 or IMP3. The relationship between nestin and PFS was independent of stage, LVSI and risk categorization but not type of UC. CONCLUSIONS: High nestin was more common in UCs with aggressive features and poor outcome. Nestin may represent a predictive biomarker for treatment selection for patients previously considered to be lower risk and a candidate for no or radiation-based adjuvant therapy, and compliment ER/PR testing.


Subject(s)
Adenocarcinoma/chemistry , Biomarkers, Tumor/analysis , Carcinosarcoma/chemistry , Nestin/analysis , Uterine Neoplasms/chemistry , Uterine Neoplasms/pathology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Carcinosarcoma/pathology , Carcinosarcoma/therapy , Disease-Free Survival , Female , Gene Expression , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Nestin/genetics , PTEN Phosphohydrolase/analysis , Peptides/analysis , Peptides/genetics , Predictive Value of Tests , RNA, Messenger/analysis , RNA-Binding Proteins/genetics , Receptors, Estrogen/analysis , Receptors, Estrogen/genetics , Receptors, Progesterone/analysis , Receptors, Progesterone/genetics , Risk Assessment , Survival Rate , Tissue Array Analysis , Trefoil Factor-3 , Tumor Suppressor Protein p53/genetics , Uterine Neoplasms/therapy
14.
J Cell Biochem ; 117(6): 1279-87, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26774024

ABSTRACT

The transforming growth factor-ß (TGF-ß) is a family of structurally related proteins that comprises of TGF-ß, activins/inhibins, and bone morphogenic proteins (BMPs). Members of the TGF-ß family control numerous cellular functions including proliferation, apoptosis, differentiation, epithelial-mesenchymal transition (EMT), and migration. The first identified member, TGF-ß is implicated in several human diseases, such as vascular diseases, autoimmune disorders, and carcinogenesis. Activation of the TGF-ß receptor by its ligands induces the phosphorylation of serine/threonine residues and triggers phosphorylation of the intracellular effectors, SMADs. Upon activation, SMAD proteins translocate to the nucleus and induce transcription of their target genes, regulating several cellular functions. TGF-ß dysregulation has been implicated in carcinogenesis. In early stages of cancer, TGF-ß exhibits tumor suppressive effects by inhibiting cell cycle progression and promoting apoptosis. However, in late stages TGF-ß exerts tumor promoting effects, increasing tumor invasiveness, and metastasis. Furthermore, the TGF-ß signaling pathway communicates with other signaling pathways in a synergistic or antagonistic manner and regulates cellular functions. Elevated TGF-ß activity has been associated with poor clinical outcome. Given the pivotal role of TGF-ß in tumor progression, this pathway is an attractive target for cancer therapy. Several therapeutic tools such as TGF-ß antibodies, antisense oligonucleotides, and small molecules inhibitors of TGF-ß receptor-1 (TGF-ßR1) have shown immense potential to inhibit TGF-ß signaling. Finally, in the interest of developing future therapies, further studies are warranted to identify novel points of convergence of TGF-ß with other signaling pathways and oncogenic factors in the tumor microenvironment.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Antineoplastic Agents , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/drug therapy , Phosphorylation , Prognosis , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
15.
Acta Pol Pharm ; 72(5): 829-42, 2015.
Article in English | MEDLINE | ID: mdl-26665389

ABSTRACT

Hispidulin is well-known natural bioactive flavone on behalf of its pharmacological aspects. This review contains data on isolation, synthetic methodology, pharmacokinetics and bioactivities of hispidulin. The article provides a critical assessment of present knowledge about hispidulin with some clear conclusions, perspectives and directions for future research in potential applications.


Subject(s)
Flavones/pharmacology , Animals , Anticonvulsants/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Flavones/chemical synthesis , Flavones/isolation & purification , Flavones/pharmacokinetics , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Osteoclasts/drug effects
16.
J Cell Biochem ; 116(8): 1797-805, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25683036

ABSTRACT

Transforming growth factor-ß (TGF-ß), regulates cell proliferation, angiogenesis, metastasis, and is an inducer of epithelial-mesenchymal transition (EMT). Cancer cells exhibit activated TGF-ß/SMAD signaling pathway and its inhibition is an attractive strategy for cancer treatment. The Chinese Herbs Scutellaria baicalensis (SB) and Fritillaria cirrhosa (FC) have been shown to be beneficial to cancer patients, but the mechanisms by which the extracts of two herbs elicit the beneficial effects are unclear. In this study, we have used human endometrial cancer cells to assess the anticancer efficacy of SB and FC on TGF-ß signaling pathway components. SB and FC treatment of cancer cells resulted in a significant decrease in expression of TGF-ß isoforms, TGF-ß receptors, and SMADs. Both herbs effectively inhibited basal and TGF-ß1-induced cancer cell proliferation and invasion, which was accompanied with abrogation of Snail, Slug, matrix metalloproteinases (MMPs), αvß3 integrin, focal adhesion kinase (FAK), and p-FAK expression. An inhibitor of TGF-ßRI blocked TGF-ß1-induced cell invasion and significantly diminished antitumor effects of SB and FC. These results suggest that SB and FC block endometrial cancer growth by downregulating TGF-ß/SMAD signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Endometrial Neoplasms/metabolism , Fritillaria/chemistry , Plant Extracts/pharmacology , Scutellaria baicalensis/chemistry , Signal Transduction/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Transforming Growth Factor beta/metabolism
17.
Mol Carcinog ; 54(5): 368-78, 2015 May.
Article in English | MEDLINE | ID: mdl-24249479

ABSTRACT

The herbs Scutellaria baicalensis (SB) and Fritillaria cirrhosa (FC) are widely used in Chinese medicine to treat several aliments and as an adjuvant to chemotherapy of lung cancer. No information is available regarding the two herbs' influence on ovarian and endometrial cancer. To fill this data gap we compared cell growth responses to SB and FC in ovarian and endometrial cancer cell lines. Dose-dependent cell growth inhibition was observed following higher doses in all cell lines while lower doses stimulated growth in only endometrial cell lines. Higher doses of SB and FC significantly decreased cell growth on soft agar and decreased the invasive potential of cancer cells. Treatment of cells with both herbs resulted in activation of caspase-3, G0 /G1 phase cell cycle arrest, downregulation of cyclins D1 and D3 and induction of p27. Both herbs decreased NFκB DNA binding, reduced expression of phosphorylated IκBα, abrogated NFκB activation, and downregulated NFκB-regulated metastasis-promoting proteins in cancer cells. Furthermore, knockdown of NFκB attenuated SB- and FC-induced cell growth inhibition. These results suggest that inhibition of NFκB activation may be an important mechanism for growth suppression by SB and FC. Data indicate that these herbs may represent a new source of agents for NFκB inhibition in cancer therapy.


Subject(s)
Cell Proliferation/drug effects , Drugs, Chinese Herbal/pharmacology , Endometrial Neoplasms/pathology , Fritillaria/chemistry , NF-kappa B/antagonists & inhibitors , Ovarian Neoplasms/pathology , Phytotherapy , Scutellaria baicalensis/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Movement/drug effects , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , RNA, Small Interfering/genetics , Tumor Cells, Cultured
18.
Cancer Prev Res (Phila) ; 7(10): 1045-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070663

ABSTRACT

Increased expression of TGFß isoforms in human endometrial cancer correlates with decreased survival and poor prognosis. Progesterone has been shown to exert a chemoprotective effect against endometrial cancer, and previous animal models have suggested that these effects are accompanied by changes in TGFß. The goal of this study was to characterize the effect of progesterone on TGFß signaling pathway components and on TGFß-induced protumorigenic activities in endometrial cancer cell lines. Progesterone significantly decreased expression of three TGFß isoforms at 72 hours after treatment except for TGFß2 in HEC-1B and TGFß3 in Ishikawa cells. Progesterone treatment for 120 hours attenuated expression of the three isoforms in all cell lines. Progesterone exposure for 72 hours reduced expression of TGFß receptors in HEC-1B cells and all but TGFßR1 in Ishikawa cells. Progesterone reduced TGFßR3 expression in RL-95 cells at 72 hours, but TGFßR1 and ßR2 expression levels were not affected by progesterone at any time point. SMAD2/3 and pSMAD2/3 were substantially reduced at 72 hours in all cell lines. SMAD4 expression was reduced in RL-95 cells at 24 hours and in HEC-1B and Ishikawa cells at 72 hours following progesterone treatment. Furthermore, progesterone effectively inhibited basal and TGFß1-induced cancer cell viability and invasion, which was accompanied by increased E-cadherin and decreased vimentin expression. An inhibitor of TGFßRI blocked TGFß1-induced effects on cell viability and invasion and attenuated antitumor effects of progesterone. These results suggest that downregulation of TGFß signaling is a key mechanism underlying progesterone inhibition of endometrial cancer growth.


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Progesterone/metabolism , Transforming Growth Factor beta/metabolism , Antigens, CD , Cadherins/metabolism , Cell Line, Tumor , Cell Survival , Culture Media , Culture Media, Conditioned/chemistry , Epithelial-Mesenchymal Transition , Female , Humans , Ligands , Microscopy, Fluorescence , Neoplasm Invasiveness , Signal Transduction , Smad4 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism
19.
BMJ Case Rep ; 20142014 Jun 26.
Article in English | MEDLINE | ID: mdl-24969070

ABSTRACT

A 52-year-old Caucasian man presented with sudden onset of difficulty in moving his tongue to the left with preceding left-sided headache with no neck pain. Earlier, he had self-limiting chest infection without rashes or tonsillar enlargement. His medical and surgical history was unremarkable with no recent trauma. Oral examination revealed difficulty in protruding his tongue to the left with muscle bulk loss and fasciculation on the same side, suggesting left hypoglossal nerve palsy. Examination of the rest of the cranial nerves and nervous system was normal. The patient's oropharyngeal and laryngeal examination was unremarkable with no cervical lymphadenopathy. He had normal laboratory investigations and cerebrospinal fluid examination. Extensive imaging of the head, neck and chest failed to reveal any pathology. Further review by an otorhinologist and rheumatologist ruled out any other underlying pathology. He made a good recovery without treatment. English literature search revealed very few cases of idiopathic, transient, unilateral hypoglossal nerve palsy.


Subject(s)
Hypoglossal Nerve Diseases/diagnosis , Hypoglossal Nerve , Tongue Diseases/diagnosis , Tongue/innervation , Humans , Male , Middle Aged , Paralysis/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...