Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Reprod Sci ; 29(5): 1560-1576, 2022 05.
Article in English | MEDLINE | ID: mdl-34424529

ABSTRACT

The oviduct is a site for early reproductive events including gamete maturation, fertilization, and early embryo development. Secretory cells lining the oviduct lumen synthesize and secrete proteins that interact with gametes and developing embryos. Although previous studies have identified some of the secretory proteins in the oviduct, however, knowledge and their precise specific functions in the oviduct are poorly understood. In this study, by using proteomic approach, we identified a secretory protein, Peroxiredoxin 6 (PRDX6), and evaluated its role in mediating early pregnancy events, fertilization, and embryo development in rabbit oviduct. The expression of PRDX6 was significantly higher in ampulla and isthmus sections of the oviduct in mated animal groups compared to non-mated controls. Furthermore, significant reduction in number of embryos recovered from PRDX6 siRNA-transfected oviductal horn was observed compared to the control contralateral horn. Moreover, in animals receiving PRDX6 siRNA in their oviductal horn, the number of implanted blastocysts was significantly less in the uterus as observed on day 9 post-coital (p.c.). Further, during embryo-rabbit oviduct epithelial cell (ROEC) co-culture, siRNA-mediated PRDX6 silencing attenuated the early embryonic development. Mechanistically, increased levels of ROS and expression of oxidative stress- and inflammation-related proteins were found in PRDX6 siRNA-treated ROEC cells as compared to control cells, implicating that ablation of PRDX6 in the oviduct creates a stress-induced micro-environment detrimental to early embryonic development in oviduct. Taken together, our data suggest that PRDX6 maintains an optimal micro-environment conducive to successful embryo development and can be considered as a candidate to evaluate its therapeutic potential in IVF strategies.


Subject(s)
Embryonic Development , Fertilization , Peroxiredoxin VI , Proteomics , Animals , Fallopian Tubes , Female , Oviducts/metabolism , Peroxiredoxin VI/metabolism , Pregnancy , Proteins/metabolism , RNA, Small Interfering/metabolism , Rabbits
2.
Cytokine ; 148: 155700, 2021 12.
Article in English | MEDLINE | ID: mdl-34560609

ABSTRACT

Transforming growth factor (TGFß) is known to play a major role in establishment and maintenance of endometriosis as reported by our group earlier, the underlying mechanism remains to be explored. We deciphered the involvement of TAK1 in TGFß1- induced cellular responses and delineated the signaling mechanism in human endometriotic cells. The endometriotic cells showed elevated expression of TGFß1 signaling-effector molecules. TGFß1 exposure to endometriotic cells induced the expression of the downstream target molecules indicating that TGFß1 is implicated in the commencement ofTAK1/NFκB-p65/Smad7 cascade. The silencing of TAK1 in endometriotic cells attenuated the TGFß1 -induced NFκB transcriptional activation and nuclear translocation of NFκB-p65 subunit. The pharmacological inhibition of NFκB by QNZ or knockdown of TAK1 reduced the expression of Smad7 and Cox2. The knockdown of TAK1 in endometriotic cells showed G1 phase cell-cycle arrest and showed low BrdU-incorporation in the presence of TGFß1. The inhibition of TAK1 attenuated the TGFß1 signaling activation indicating that TAK1 is a crucial mediator for TGFß1 action in endometriotic cells. The exposure of endometriotic cells to TAK1 inhibitor, celastrol caused activation of caspase-3 and -9 that led to PARP cleavage and induced apoptosis. Simultaneously, autophagy occurred in celastrol-treated and TAK1-silenced cells as was evidenced by the formation of autophagosome and the increased expression of autophagic markers. Thus, TAK1 activation appears to protect the growth of endometriotic cells by suppressing the cell death process. Overall, our study provided the evidence that of TAK1 significant in the endometriotic cell regulation and mediates a functional cross-talk between TGFß1 and NFκB-p65 that promotes the growth and inflammatory response in endometriotic cells.


Subject(s)
Autophagy , Endometriosis/metabolism , Endometriosis/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Signal Transduction , Smad7 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Endometrium/pathology , Female , G1 Phase/drug effects , Gene Knockdown Techniques , Humans , Models, Biological , Pentacyclic Triterpenes/pharmacology , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism
3.
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
5.
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
6.
Reproduction ; 158(1): 47-59, 2019 07.
Article in English | MEDLINE | ID: mdl-30952110

ABSTRACT

Microtubule (MT) dynamics plays a crucial role in fertilization and early embryonic development; however its involvement in uterus during embryo implantation remains unclear. Herein, we report the effect of microtubule depolymerization during embryo implantation in BALB/c mice. Intrauterine treatment with depolymerizing agent nocodazole at pre-implantation phase (D4, 07:00 h) in mice resulted into mitigation in receptivity markers viz. LIF, HoxA10, Integrin-ß3, IHH, WNT4 and led to pregnancy failure. MT depolymerization in endometrial epithelial cells (EECs) also inhibited the blastocyst attachment and the adhesion. The decreased expression of MT polymerization-related proteins TPPP and α/ß-tubulin in luminal and glandular epithelial cells along with the alteration in morphology of pinopodes in the luminal epithelium was observed in nocodazole receiving uteri. Nocodazole treatment also led to increased intracellular Ca+2 levels in EECs, which indicated that altered Ca+2 homeostasis might be responsible for implantation failure. Microtubule depolymerization inhibited WNT4 and Fz-2 interaction, thereby suppressing the downstream WNT4/CaMKIIα signaling cascades calmodulin and calcineurin which led to attenuation of NF-κB transcriptional promoter activity in EECs. MT depolymerization or CaMKIIα knockdown inhibited the transcription factor NFAT and NF-κB expression along with reduced secretion of prostaglandins PGE2 and PGF2α in mouse EECs. Overall, MT depolymerization impaired the WNT4/CaMKIIα signaling and suppressed the secretion of PGE2 and PGF2α in EECs which may be responsible for implantation failure in mice.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Embryo Implantation , Embryonic Development , Endometrium/pathology , Microtubules/pathology , Uterus/pathology , Wnt4 Protein/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Mice , Mice, Inbred BALB C , Microtubules/drug effects , Microtubules/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nocodazole/pharmacology , Pregnancy , Signal Transduction , Tubulin Modulators/pharmacology , Uterus/drug effects , Uterus/metabolism , Wnt4 Protein/genetics
7.
J Mol Med (Berl) ; 97(4): 509-522, 2019 04.
Article in English | MEDLINE | ID: mdl-30729278

ABSTRACT

Decidualization of endometrial stromal cells is the pre-requisite for the embryo implantation and establishment of pregnancy. Although known to be regulated by several factors, the process of regulation of decidualization by miRNAs is largely unknown. Previous reports suggest that the upregulated expression of miR-145 is associated with repeated implantation failure. The current study was aimed to identify and validate the role of miR-145 in regulating stromal cell decidualization and the mechanism involved therein. Expression of miR-145 was found to be downregulated during the decidualization period of early pregnancy and also in artificially induced decidualization in rat uterus. During in vitro decidualization in rat endometrial stromal cells (ESCs), the overexpression of mimic miR-145 attenuated the progression of decidualization. Biochemical marker alkaline phosphatase and protein markers (insulin-like growth factor binding protein, cyclin D3) were also suppressed in miR-145 mimic-transfected cells as compared to normal decidualized cells. Bioinformatic analysis and luciferase reporter assay confirmed that Smad1 is the direct target of miR-145. Differentiation of ESCs was inhibited in miR-145 mimic-transfected cells which occurred via downregulating the target Smad1 along with its downstream p-Smad1/5/8 and Wnt-4. Pre-treatment of ESCs with Smad1 siRNA resulted in downregulated expression of p-Smad1/5/8, Wnt-4, Cox-2, and VEGF. In addition, miR-145 overexpression resulted in the loss of angiogenic factors Cox-2, MMP-9, and VEGF, indicating suppression of the process of angiogenesis. Migration of human umbilical vein endothelial cells was also attenuated in the presence of conditioned media obtained from miR-145-transfected decidualizing cells. In conclusion, the study demonstrated the role of miR-145 in regulation of progression of decidualization which is mediated through inhibition of Smad1. KEY MESSAGES: MiR-145 expression is downregulated during decidualization in the rat uterus. Overexpression of miR-145 inhibited the decidualization progression. MiR-145 suppressed the migration and invasion of HUVECs. MiR-145 downregulated Smad1 which suppresses Smad1/5/8, Wnt-4, MMP-9, Cox-2, and VEGF.


Subject(s)
Decidua/cytology , Endometrium/cytology , MicroRNAs/genetics , Smad1 Protein/genetics , Stromal Cells/cytology , Animals , Cell Proliferation , Decidua/metabolism , Down-Regulation , Embryo Implantation , Endometrium/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Pregnancy , Rats , Rats, Sprague-Dawley , Stromal Cells/metabolism , Up-Regulation
8.
J Endocrinol ; 240(3): 417-429, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30667362

ABSTRACT

Embryo implantation and decidualization are critical events that occur during early pregnancy. Decidualization is synchronized by the crosstalk of progesterone and the cAMP signaling pathway. Previously, we confirmed the role of TPPP3 during embryo implantation in mice, but the underlying role and mechanism of TPPP3 in decidualization has not yet been understood. The current study was aimed to investigate the role of TPPP3 in decidualization in vivo and in vitro. For in vivo experiments, decidual reaction was artificially induced in the uteri of BALB/c mice. TPPP3 was found to be highly expressed during decidualization, whereas in the uteri receiving TPPP3 siRNA, decidualization was suppressed and the expression of ß-catenin and decidual marker prolactin was reduced. In human endometrium, TPPP3 protein was found to be predominantly expressed in the mid-secretory phase (LH+7). In the primary culture of human endometrial stromal cells (hESCs), TPPP3 siRNA knockdown inhibited stromal-to-decidual cell transition and decreased the expression of the decidualization markers prolactin and IGFBP-1. Immunofluorescence and immunoblotting experiments revealed that TPPP3 siRNA knockdown suppressed the expression of ß-catenin, NF-κB and COX-2 in hESCs during decidualization. TPPP3 inhibition also decreased NF-kB nuclear accumulation in hESCs and suppressed NF-κB transcriptional promoter activity. COX-2 expression was significantly decreased in the presence of a selective NF-kB inhibitor (QNZ) implicating that NF-kB is involved in COX-2 expression in hESCs undergoing decidualization. TUNEL assay and FACS analysis revealed that TPPP3 knockdown induced apoptosis and caused loss of mitochondrial membrane potential in hESCs. The study suggested that TPPP3 plays a significant role in decidualization and its inhibition leads to the suppression of ß-catenin/NF-κB/COX-2 signaling along with the induction of mitochondria-dependent apoptosis.


Subject(s)
Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Decidua/metabolism , Embryo Implantation , Endometrium/metabolism , Signal Transduction , Stromal Cells/metabolism , Adult , Animals , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytoskeletal Proteins/genetics , Decidua/cytology , Endometrium/cytology , Female , Humans , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/metabolism , Pregnancy , RNA Interference , Stromal Cells/cytology , Uterus/cytology , Uterus/metabolism , beta Catenin/genetics , beta Catenin/metabolism
9.
Free Radic Biol Med ; 129: 582-599, 2018 12.
Article in English | MEDLINE | ID: mdl-30347228

ABSTRACT

Hh/Gli1 cascade as well as Gsk3ß-Gli1 crosstalk play crucial role in estrogen-dependent progression of endometrial hyperplasia (EH). However, the underlying mechanisms involved in progression of disease still remain unclear. In the present study, we explored the role of Hh signaling in protection of endometrial hyperplasial cells against oxidative stress and the underlying mechanism involved therein. EH cells were found to be more resistant towards H2O2-induced oxidative stress (IC50: ~ 3×) as compared with normal endometrial cells. Estrogen (E2) pre-treatment followed by cytotoxic dose of H2O2, almost rescued the EH cells from apoptosis and caused the increased expression of downstream Shh signaling molecules i.e., Smo, Ptch and Gli1. Whereas pretreatment with cyclopamine was not able to curtail H2O2-induced effects indicating that estrogen protects these cells via activation of Shh pathway. Further, H2O2-induced ROS and lipid peroxidation alongwith decreased activities of antioxidant enzymes glutathione peroxidase and superoxide dismutase were found to be reversed in EH cells pre-exposed to E2 or rShh. The rShh suppressed H2O2-induced cell death and caused attenuation of mitochondrial apoptotic mediators and prevented disruption in mitochondrial morphology and mitochondrial membrane potential in EH cells. The functional blockage of signaling by Shh siRNA or Gli1siRNA led to significantly increased expression of mitochondrial fission protein dynamin-like GTPase (Drp1). The H2O2-treated EH cells showed diminished Gli1 and increased Drp1 expression, concurrent with reduced p-Drp1-(serine637). Whereas rShh pre-treated EH cells presented normal mitochondrial dynamics with dense, long networks of mitochondria alongwith nuclear accumulation of Gli1 and the decreased expression of Drp1. Overall, our results implicated that Shh signaling modulates antioxidant defense system and stabilizes mitochondrial dynamics by suppressing Drp1 protein which maintains survival of EH cells against oxidative stress.


Subject(s)
Endometrial Hyperplasia/genetics , Epithelial Cells/metabolism , GTP Phosphohydrolases/genetics , Hedgehog Proteins/genetics , Microtubule-Associated Proteins/genetics , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/genetics , Zinc Finger Protein GLI1/genetics , Adult , Animals , Case-Control Studies , Disease Progression , Dynamins , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrial Hyperplasia/surgery , Endometrium/metabolism , Endometrium/pathology , Endometrium/surgery , Epithelial Cells/drug effects , Epithelial Cells/pathology , Estrogens/pharmacology , Female , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Hedgehog Proteins/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Hysterectomy , Lipid Peroxidation/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism
10.
Biol Reprod ; 99(5): 982-999, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29901777

ABSTRACT

Tubulin polymerization promoting protein 3 (TPPP3) is known to be expressed in the endometrium in a cyclic manner, and its functional role in the physiology of implantation remains unknown. Here we demonstrate a novel function of TPPP3 during the window of implantation and in the establishment of pregnancy using a mouse model. The increased protein expression of TPPP3 and ß-catenin during peri-implantation period, i.e. D5 (receptive phase, 0800 h), was observed as compared to that on D1 (nonreceptive phase, 0800 h). SiRNATPPP3-mediated knockdown of uterine TPPP3 resulted in implantation failure and inhibited the expression of receptivity markers: LIF, Integrin-ß3, IHH, and Wnt4. TPPP3 silencing in mouse endometrial epithelial cells also prevented blastocyst attachment and the adhesion reaction. In delayed implantation experiment, expression of TPPP3 was increased in active implantation group (E2 + P4) compared to delayed implantation group (P4). The increased expression of TPPP3 in E2 + P4-treated Ishikawa cells compared to vehicle or P4 or E2 alone-treated Ishikawa cells also revealed its upregulation by E2. The suppression of ß-catenin in uterus under the condition of transient knockdown of TPPP3 and the co-immunoprecipitation experiment revealed that regulation of ß-catenin was mediated via TPPP3 during implantation. Additionally, in order to gain insight into TPPP3 collaborators, we identified TPPP3 interacting proteins by nanoLC-MS analysis in mouse uterus which might be involved during implantation. In conclusion, our study suggests that TPPP3 is important for embryo implantation and for the establishment of early pregnancy through modulation of ß-catenin.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Embryo Implantation/genetics , Embryo Implantation/physiology , Uterus/metabolism , beta Catenin/metabolism , Animals , Blastocyst , Cell Line, Tumor , Estradiol/pharmacology , Female , Gene Knockdown Techniques , Mice , Mice, Inbred BALB C , Pregnancy , Pseudopregnancy/genetics , Pseudopregnancy/metabolism
11.
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
12.
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
14.
Sci Rep ; 7(1): 6557, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747625

ABSTRACT

The present study was undertaken to explore the functional involvement of Hh signaling and its regulatory mechanism in endometrial hyperplasia. Differential expression of Hh signaling molecules i.e., Ihh, Shh, Gli1 or Gsk3ß was observed in endometrial hyperplasial (EH) cells as compared to normal endometrial cells. Estradiol induced the expression of Hh signaling molecules and attenuated the expression of Gsk3ß whereas anti-estrogen (K1) or progestin (MPA) suppressed these effects in EH cells. Cyclopamine treatment or Gli1 siRNA knockdown suppressed the growth of EH cells and reduced the expression of proliferative markers. Estradiol also induced the nuclear translocation of Gli1 which was suppressed by both MPA and K1 in EH cells. While exploring non-canonical mechanism, LY-294002 (Gsk3ß activator) caused a decrease in Gli1 expression indicating the involvement of Gsk3ß in Gli1 regulation. Further, Gsk3ß silencing promoted the expression and nuclear translocation of Gli1 demonstrating that Gsk3ß serves as a negative kinase regulator of Gli1 in EH cells. Similar attenuation of Hh signaling molecules was observed in rats with uterine hyperplasia undergoing anti-estrogen treatment. The study suggested that Hh/Gli1 cascade (canonical pathway) as well as Gsk3ß-Gli1 crosstalk (non-canonical pathway) play crucial role in estrogen-dependent cell proliferation in endometrial hyperplasia.


Subject(s)
Cell Proliferation , Endometrial Hyperplasia/physiopathology , Estrogens/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Zinc Finger Protein GLI1/metabolism , Cells, Cultured , Female , Humans
15.
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
16.
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
17.
Mol Cell Endocrinol ; 448: 41-54, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28288902

ABSTRACT

A series of new 6H-benzofuro[3, 2-c]chromenes (BFC, pterocarpans) with structure-activity relationships were investigated for their potential use in osteoporosis treatment. One of the BFCs 3-piperidylethoxypterocarpan 20 promotes osteoblast differentiation and mineralization at a dose as low as 1 pM via activation of ER/P38MAPK/BMP-2 pathway. When evaluated for in-vivo osteogenic activity in female Sprague-Dawley rats, BFC 20 increased bone mineral density and new bone formation, compared with control at 1.0 and 10.0 mg/kg/body weight by oral gavage for 30 days. The compound was devoid of any uterotrophic effect and led to the new bone formation in adult ovariectomized osteopenic rats. BFC 20 compound also inhibited bone resorption by reducing Ovx induced increase in urinary CTx, thus exhibiting both bone anabolic and anti-catabolic action. Finally, BFC 20 treatment to Ovx rats led to improved trabecular microarchitectural restoration and exhibited therapeutic potential as a dual acting anti-osteoporotic agent for the management of osteoporosis.


Subject(s)
Anabolic Agents/therapeutic use , Bone Diseases, Metabolic/drug therapy , Cancellous Bone/pathology , Ovariectomy , Piperidines/therapeutic use , Pterocarpans/therapeutic use , Alkaline Phosphatase/metabolism , Anabolic Agents/chemical synthesis , Anabolic Agents/chemistry , Anabolic Agents/pharmacology , Animals , Biomarkers/metabolism , Bone Density/drug effects , Bone Diseases, Metabolic/pathology , Bone Morphogenetic Protein 2/metabolism , Bone Remodeling/drug effects , Calcification, Physiologic/drug effects , Cancellous Bone/drug effects , Cell Differentiation/drug effects , Female , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphorylation/drug effects , Piperidines/chemical synthesis , Piperidines/chemistry , Piperidines/pharmacology , Pterocarpans/chemical synthesis , Pterocarpans/chemistry , Pterocarpans/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
18.
Reprod Sci ; 24(5): 738-752, 2017 05.
Article in English | MEDLINE | ID: mdl-27662902

ABSTRACT

The function of RHOG, a RAC1 activator, was explored in the ovary during ovarian follicular development and pathological conditions. With the help of immunoblotting and immunolocalization, we determined the expression and localization of RHOG in normal (estrous cycle) and polycystic ovaries using Sprague Dawley (SD) rat model. Employing polymerase chain reaction and flow cytometry, we analyzed the transcript and expression levels of downstream molecules of RHOG, DOCK1, and RAC1 in the polycystic ovarian syndrome (PCOS) ovary along with normal antral follicular theca and granulosa cells after dehydroepiandrosterone (DHEA) supplementation. The effect of RHOG knockdown on DOCK1, VAV, and RAC1 expression was evaluated in the human ovarian cells (SKOV3), theca cells, and granulosa cells from SD rats with the help of flow cytometry. Oocyte at secondary follicles along with stromal cells showed optimal expression of RHOG. Immunoblotting of RHOG revealed its maximum expression at diestrus and proestrus, which was downregulated at estrus stage. Mild immunostaining of RHOG was also present in the theca and granulosa cells of the secondary and antral follicles. Polycystic ovary exhibited weak immunostaining for RHOG and that was corroborated by immunoblotting-based investigations. RHOG effectors DOCK1 and ELMO1 were found reduced in the ovary in PCOS condition/DHEA. RHOG silencing reduced the expression of DOCK1 and RAC1 in the theca and granulosa cells from SD rat antral follicles and that was mirrored in the human ovarian cells. Collectively, RHOG can mediate signaling through downstream effectors DOCK1 and RAC1 during ovarian follicular development (theca and granulosa cells and oocyte), but DHEA downregulated them in the PCOS ovary.


Subject(s)
GTP Phosphohydrolases/metabolism , Ovarian Follicle/metabolism , Polycystic Ovary Syndrome/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Line , Dehydroepiandrosterone , Disease Models, Animal , Estrous Cycle , Female , Humans , Ovarian Follicle/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology , Puberty , Rats , Rats, Sprague-Dawley , Signal Transduction
19.
Bioconjug Chem ; 27(4): 961-72, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26999669

ABSTRACT

The synthesis of estradiol based bivalent ligand [(EST)2DT] is reported and its potential for targeted imaging and therapy of ER(+) tumors has been evaluated. For the purpose, ethinylestradiol was functionalized with an azidoethylamine moiety via click chemistry. The resultant derivative was reacted in a bivalent mode with DTPA-dianhydride to form the multicoordinate chelating agent, (EST)2DT which displayed capability to bind (99m)Tc. The radiolabeled complex, (99m)Tc-(EST)2DT was obtained in >99% radiochemical purity and 20-48 GBq/µmol of specific activity. RBA assay revealed ∼15% binding with estrogen receptor. Evaluation of ligand on ER(+)-cell line (MCF-7) suggested enhanced and ER-mediated uptake. In vivo assays displayed early tracer accumulation in MCF-7 xenografts with tumor to muscle ratio ∼6 in 2 h and negligible uptakes in nontargeted organs. MTT assay performed on ER(+) and ER(-) cell lines displayed selective inhibition of ER(+) cancer cell growth with IC50 = 14.3 µM which was comparable to tamoxifen. The anticancer activity of the ligand is possibly due to the increase in ERß and suppression of ERα protein levels in gene transcription. The studies reveal the potential of (EST)2DT as diagnostic imaging agent with the additional benefits in therapy.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Estradiol/metabolism , Receptors, Estrogen/metabolism , Theranostic Nanomedicine , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Dimerization , Female , Humans , Ligands , Mice , Mice, Nude , Tissue Distribution
20.
Int J Biochem Cell Biol ; 70: 105-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26556313

ABSTRACT

Breast cancer remains a significant health problem due to the involvement of multiple aberrant and redundant signaling pathways in tumorigenesis and the development of resistance to the existing therapeutic agents. Therefore, the search for novel chemotherapeutic agents for effective management of breast cancer is still warranted. In an effort to develop new anti-breast cancer agents, we have synthesized and identified novel spiro-oxindole derivative G613 i.e. 5-chloro-4',5'-diphenyl-3'-(4-(2-(piperidin-1-yl) ethoxy) benzoyl) spiro[indoline-3,2'-pyrrolidin]-2-one, which has shown growth inhibitory activity in breast cancer cells. The present study was aimed to explore the mechanism of anti-tumorigenic action of this newly identified spiro-oxindole compound. Compound G613 inhibited the Mdm2-p53 interaction in breast cancer cells and tumor xenograft. It caused restoration of p53 function by activating its promoter activity, triggering its nuclear accumulation and preventing its ubiquitination and proteasomal degradation. Supportively, molecular docking studies revealed considerable homology in the docking mode of G613 and the known Mdm2 inhibitor Nutlin-3, to p53 binding pocket of Mdm2. The activation of p53 led to upregulation of p53 dependent pro-apoptotic proteins, Bax, Pumaα and Noxa and enhanced interaction of p53 with bcl2 member proteins thus triggering both transcription-dependent and transcription-independent apoptosis, respectively. Additionally, the compound decreased estrogen receptor activity through sequestration of estrogen receptor α by p53 thereby causing a decreased transcriptional activation and expression of proliferation markers. In conclusion, G613 represents a potent small-molecule inhibitor of the Mdm2-p53 interaction and can serve as a promising lead for developing a new class of anti-cancer therapy for breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Spiro Compounds/pharmacology , Aged , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Proliferation/drug effects , Chlorocebus aethiops , Female , HEK293 Cells , Humans , Imidazoles/pharmacology , Indoles/chemical synthesis , MCF-7 Cells , Mice , Mice, Nude , Middle Aged , Molecular Docking Simulation , Piperazines/pharmacology , Protein Binding/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Spiro Compounds/chemical synthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vero Cells , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...