Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 42: 116252, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34153643

ABSTRACT

Breast cancer is the second leading cause of cancer deaths in women with significant morbidity and mortality. Present study describes design, synthesis and detailed pharmacology of indole derivatives exhibiting remarkable broad spectrum antiproliferative activity against breast cancer cells. Detailed mechanistic evaluations confirmed induction of G0/G1 arrest, apoptosis induction, loss of mitochondrial integrity, enhanced ROS generation, autophagy, estrogen receptor ß-transactivation and increased tubulin polymerization. In in-vivo efficacy studies in rodent model, these indole derivatives induced significant regression in mice mammary tumour on 21 days daily oral dose. Moreover, compounds 19 and 23 were safe in Swiss albino mice in safety studies. These diarylindoles may further be optimized for better efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Drug Design , Indoles/pharmacology , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Indoles/chemical synthesis , Indoles/chemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
2.
J Nutr Biochem ; 70: 215-226, 2019 08.
Article in English | MEDLINE | ID: mdl-31252288

ABSTRACT

Endometriosis is mainly characterized by the presence of endometrial tissue exterior to the uterus, however, the exact pathophysiology of this disease still remains uncertain. Moreover, the incidence significantly contributes to infertility among women and hence, a novel treatment for endometriosis is widely investigated. Naringenin is a plant-derived flavonoid having anti-proliferative, anti-inflammatory, and anti-angiogenic properties in chronic and metabolic diseases. The current study was planned with an objective to demonstrate the anti-endometriotic therapeutic potential of naringenin in rats and to examine its impact on various cellular aspects with a view to define the mechanism involved. The endometrial lesion volumes, weight, serum TNF-α level and the histopathologic scores were significantly reduced in the naringenin- treated group as compared to the endometriotic control group. Naringenin ameliorated the expression of prognostic markers (TAK1, PAK1, VEGF and PCNA) involved in development and progression of endometriotic cells. Naringenin caused dose-dependent loss of mitochondrial membrane potential, induced apoptosis and inhibited proliferation in these cells. Further, a significant increase in level of Nrf2 and its downstream molecules (NQO1, HO-1) was found in endometriotic lesion, with a subsequent decrease in its repressor molecule Keap-1. Naringenin significantly modulated the expression of Nrf2 and its effector molecules downstream. It also inhibited the invasion of endometrial cells by reducing the expression of MMP-2 and MMP-9 in in-vitro primary culture. We conclude that naringenin may have a therapeutic potential in the treatment of endometriosis via induction of ROS-mediated apoptosis and its anti-invasive effects.


Subject(s)
Apoptosis , Endometriosis/drug therapy , Flavanones/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cell Movement , Disease Progression , Endometrium/drug effects , Female , Membrane Potential, Mitochondrial , Neoplasm Transplantation , Nitric Oxide/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
3.
J Steroid Biochem Mol Biol ; 178: 272-282, 2018 04.
Article in English | MEDLINE | ID: mdl-29330027

ABSTRACT

Ormeloxifene, the non-steroidal SERM contraceptive, inhibits endometrial receptivity and embryo implantation via countering nidatory estrogen. However, the molecular mechanism of ormeloxifene action responsible for its contraceptive efficacy still remains unclear. Herein, we aimed to identify the miRNAs modulated under the influence of ormeloxifene and to explore their role in endometrial receptivity and embryo implantation. By doing microRNA sequencing analysis, a total of 168 miRNAs were found to be differentially expressed in uterine tissue of ormeloxifene-treated rats, on day 5 (10:00 h) of pregnancy i.e. peri-implantation period. Out of differentially expressed miRNAs, miR-140 expression was found to be elevated in ormeloxifene administered groups and was selected for detailed investigation. In-vivo gain-of-function of miR-140 resulted in a significant reduction of implantation sites indicating its role in embryo implantation. The experiment on delayed implantation showed that estradiol caused down-regulation of miR-140. It also suppressed the attachment and outgrowth of BeWo spheroids to RL95-2 endometrial cells. In transwell migration assay, miR-140 was found to be involved in suppression of migration and invasion of endometrial epithelial cells. The ormeloxifene treatment caused up-regulation of miR-140 along with down-regulated expression of its target IGF1R in endometrial epithelial and stromal cells which also led to the suppression of downstream effectors integrin ß3 and FAK. In mimic miR-140 receiving horn, the reduced expression of IGF1R was observed along with suppressed downstream integrin ß3 and FAK similar to that observed in uteri of ormeloxifene- treated rats. Taken together, these findings suggest that ormeloxifene-induced inhibition of embryo implantation occurs via inducing miR-140 and altering its target IGF1R in rat uterus.


Subject(s)
Benzopyrans/pharmacology , Embryo Implantation/drug effects , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Receptors, Somatomedin/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Uterus/drug effects , Animals , Cell Movement , Embryo Implantation/physiology , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Somatomedin/genetics , Uterus/metabolism
4.
J Mol Endocrinol ; 60(2): 119-132, 2018 02.
Article in English | MEDLINE | ID: mdl-29273681

ABSTRACT

Our earlier studies have demonstrated the cyclic variation and also the altered expression of sorcin in endometrium during early-to-mid-secretory phase transition in women with unexplained infertility. The current study was undertaken to establish the functional role of sorcin in endometrial receptivity in mice. Results indicated that sorcin was highly expressed during the window of implantation in mice and functional blockage of sorcin caused significant reduction in number of implanted blastocyst. The receptivity markers (i.e.Integrin ß3, HBEGF, IGFBP1, WNT4 and Cyclin E)) were found to be downregulated in sorcin knocked down uterine horn on day 5 as compared to untreated horn. The reduced attachment and expansion of BeWo spheroids on RL95-2 endometrial cells with sorcin knock down, in in vitro model of endometrium-trophoblast interaction further supported these findings. Uterine sorcin expression pattern during estrous cycle and in delayed implantation mice model suggested the upregulation of sorcin by estrogen. The functional blockade of sorcin induced the intracellular Ca+2 levels in endometrial epithelial cells (EECs), which indicated that altered Ca+2 homeostasis might be responsible for implantation failure. Sorcin silencing led to significant reduction in the expression of angiogenic factor VEGF and its downstream effector molecules i.e. PI3K, Akt and NOS. The migratory and invasive properties of HUVECs were abrogated by anti-VEGF or by adding culture media from sorcin blocked EECs, which indicated that sorcin might mediate angiogenesis during implantation. Taken together, sorcin is involved in the regulation of Ca+2-mediated angiogenesis via VEGF/PI3K/Akt pathway in endometrial cells and plays a crucial role in preparing the endometrium for implantation.


Subject(s)
Calcium-Binding Proteins/metabolism , Embryo Implantation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/genetics , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Embryo Implantation/drug effects , Endometrium/cytology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrous Cycle/drug effects , Female , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Space/metabolism , Mice , Models, Biological , Pregnancy , Signal Transduction/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Trophoblasts/cytology , Up-Regulation/drug effects , Up-Regulation/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Nanomedicine ; 13(6): 1953-1963, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28457934

ABSTRACT

Survivin is up-regulated in 83% of endometrial cancer leading to resistance development. As endometrial tumor advances, it also elicits chronic inflammation characterized by increased cytokine secretion and immune cells infiltration. The present study was designed to engineer mixed micellar curcumin loaded formulation for investigating survivin down-regulation, its anti-cancer and cytokine modulatory potential against endometrial cancer Ishikawa cells. Flory-Huggins interaction parameter (χpd) was applied to predict the compatibility between curcumin and surfactant mixture. The developed and characterized formulations were used to comparatively assess hemolysis, cellular uptake, cell-viability, apoptosis, mitochondrial membrane potential loss, rhodamine accumulation and bioavailability. In-vitro cytotoxicity in Vero cells demonstrated no deleterious effects on cell population. We saw better bioavailability, significant rhodamine accumulation, changes in protein expression and modulation in TNF-α, IL-6 and IL-10 levels. In conclusion, developed formulation warrants exploring the therapeutic interventions for overcoming resistance development in endometrial cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Micelles , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/metabolism , Down-Regulation , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Humans , Immunotherapy , Membrane Potential, Mitochondrial/drug effects , Rhodamines/metabolism , Survivin
7.
J Nutr Biochem ; 44: 60-70, 2017 06.
Article in English | MEDLINE | ID: mdl-28402926

ABSTRACT

Although curcumin shows anti-proliferative and anti-inflammatory activities in various cancers, the effect of curcumin on cellular migration in endometrial adenocarcinoma cells remains to be understood. The current investigation was aimed to explore the anti-proliferative and anti-migratory effects of curcumin and its mechanism of action in endometrial cancer cells. Our in-vitro and in-vivo experimental studies showed that curcumin inhibited the proliferation of endometrial cancer cells and suppressed the tumor growth in Ishikawa xenograft mouse model. Curcumin induced ROS-mediated apoptosis in endometrial cancer cells. Curcumin suppressed the migration rate of Ishikawa and Hec-1B cells as analyzed by scratch wound assay. In transwell migration studies, knock down of Slit-2 reversed the anti-migratory effect of curcumin in these cell lines. Curcumin significantly up-regulated the expression of Slit-2 in Ishikawa, Hec-1B and primary endometrial cancer cells while it down-regulated the expression of stromal cell-derived factor-1 (SDF-1) and CXCR4 which in turn, suppressed the expression of matrix metallopeptidases (MMP) 2 and 9, thus attenuating the migration of endometrial cancer cells. In summary, we have demonstrated that curcumin has inhibitory effect on cellular migration via Slit-2 mediated down-regulation of CXCR4, SDF-1, and MMP2/MMP9 in endometrial carcinoma cells. These findings helped explore the role of Slit-2 in endometrial cancer cells.


Subject(s)
Cell Movement/drug effects , Chemokine CXCL12/metabolism , Curcumin/pharmacology , Endometrial Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, CXCR4/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemokine CXCL12/genetics , Chlorocebus aethiops , Down-Regulation , Endometrial Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Signal Transduction , Vero Cells , Xenograft Model Antitumor Assays
8.
J Chem Inf Model ; 57(4): 815-825, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28322559

ABSTRACT

Trypansomatids maintain their redox balance by the trypanothione-based redox system, enzymes of which exhibit differences from mammalian homologues. γ-Glutamylcysteine synthetase (Gcs) is an essential enzyme in this pathway that performs the first and rate-limiting step. l-Buthionine-(S,R)-sulfoximine (BSO), a specific inhibitor of Gcs, induces toxicity in hosts infected with Trypanosoma brucei, underlining the need for novel Gcs inhibitors. The present study reports identification of Leishmania donovani Gcs (LdGcs) inhibitors using computational approaches and their experimental validation. Analysis of inhibitor-LdGcs complexes shows modifications that could result in increased efficacy of these compounds.


Subject(s)
Dipeptides/antagonists & inhibitors , Dipeptides/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Leishmania donovani/enzymology , Molecular Dynamics Simulation , Amino Acid Sequence , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Dipeptides/chemistry , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Leishmania donovani/drug effects , Protein Conformation , User-Computer Interface
9.
J Steroid Biochem Mol Biol ; 154: 130-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26241029

ABSTRACT

The oviduct plays a crucial role in female reproduction by regulating gamete transport, providing a specific microenvironment for fertilization and early embryonic development. Cyclooxygenase (COX)-derived prostaglandins play essential role in carrying out these oviduct-specific functions. Estrogen upregulates COX-2 expression in rat oviduct; however, the mechanisms responsible for regulation of COX-2 expression in rat oviductal epithelial cells (OECs) remain unclear. In the present study, we proposed that estrogen induces COX-2 expression via G-protein coupled receptor i.e., GPR30 in OECs. To investigate this hypothesis, we examined the effects of E2-BSA, ICI 182,780, GPR30 agonist and GPR30 antagonist on COX-2 expression and explored potential signaling pathway leading to COX-2 expression. Co-localization experiments revealed GPR30 to be primarily located in the peri-nuclear space, which was also the site of E2-BSA-fluorescein isothiocyanate (E2-BSA-FITC) binding. The E2-BSA induced-COX-2 and prostaglandin release were subjected to regulation by both EGFR and PI3K signaling as inhibitors of c-Src kinase (PP2), EGFR (EGFR inhibitor) and PI-3 kinase (LY294002) attenuated E2-BSA mediated effect. These results suggest that EGFR transactivation leading to activation of PI-3K/Akt pathway participates in COX-2 expression in rat OECs. Interestingly, E2-BSA induced COX-2 expression and subsequent prostaglandin release were abolished by NF-κB inhibitor. In addition, E2-BSA induced the nuclear translocation of p65-NF-κB and up-regulated the NF-κB promoter activity in rat OECs. Taken together, results demonstrated that E2-BSA induced the COX-2 expression and consequent PGE2 and PGF2α release in rat OECs. These effects are mediated through GPR30-derived EGFR transactivation and PI-3K/Akt cascade leading to NF-κB activation.


Subject(s)
Cyclooxygenase 2/metabolism , ErbB Receptors/metabolism , Oviducts/enzymology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , src-Family Kinases/metabolism , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Oviducts/cytology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
10.
PLoS One ; 9(11): e111687, 2014.
Article in English | MEDLINE | ID: mdl-25405865

ABSTRACT

BACKGROUND: Compromised receptivity of the endometrium is a major cause of unexplained infertility, implantation failure and subclinical pregnancy loss. In order to investigate the changes in endometrial protein profile as a cause of unexplained infertility, the current study was undertaken to analyze the differentially expressed proteins of endometrium from early-secretory (LH+2) to mid-secretory phase (LH+7), in women with unexplained infertility. METHODS: 2-D gel electrophoresis was performed to analyze the proteomic changes between early- (n = 8) and mid-secretory (n = 8) phase endometrium of women with unexplained infertility. The differentially expressed protein spots were identified by LC-MS analysis and validated by immunoblotting and immuno-histochemical analysis in early- (n = 4) and mid-secretory (n = 4) phase endometrium of infertile women. Validated proteins were also analyzed in early- (n = 4) and mid-secretory (n = 4) phase endometrium of fertile women. RESULTS: Nine proteins were found to be differentially expressed between early- and mid- secretory phases of endometrium of infertile women. The expression of Ras-related protein Rap-1b, Protein disulfide isomerase A3, Apolipoprotein-A1 (Apo-A1), Cofilin-1 and RAN GTP-binding nuclear protein (Ran) were found to be significantly increased, whereas, Tubulin polymerization promoting protein family member 3, Superoxide dismutase [Cu-Zn], Sorcin, and Proteasome subunit alpha type-5 were significantly decreased in mid- secretory phase endometrium of infertile women as compared to early-secretory phase endometrium of infertile women. Validation of 4 proteins viz. Sorcin, Cofilin-1, Apo-A1 and Ran were performed in separate endometrial biopsy samples from infertile women. The up-regulated expression of Sorcin and down-regulated expression of Cofilin-1 and Apolipoprotein-A1, were observed in mid-secretory phase as compared to early-secretory phase in case of fertile women. CONCLUSIONS: De-regulation of the expression of Sorcin, Cofilin-1, Apo-A1 and Ran, during early- to mid-secretory phase may have physiological significance and it may be one of the causes for altered differentiation and/or maturation of endometrium, in women with unexplained infertility.


Subject(s)
Endometrium/metabolism , Infertility, Female/metabolism , Luteal Phase/metabolism , Adult , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Case-Control Studies , Cofilin 1/genetics , Cofilin 1/metabolism , Female , Humans , Infertility, Female/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...