Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cytokine ; 148: 155700, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34560609

RESUMEN

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.


Asunto(s)
Autofagia , Endometriosis/metabolismo , Endometriosis/patología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , FN-kappa B/metabolismo , Transducción de Señal , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Endometrio/patología , Femenino , Fase G1/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Triterpenos Pentacíclicos/farmacología , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
2.
Bioorg Med Chem ; 42: 116252, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34153643

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Indoles/farmacología , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Indoles/síntesis química , Indoles/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Polimerizacion/efectos de los fármacos , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química
3.
Reproduction ; 158(1): 47-59, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30952110

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Implantación del Embrión , Desarrollo Embrionario , Endometrio/patología , Microtúbulos/patología , Útero/patología , Proteína Wnt4/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nocodazol/farmacología , Embarazo , Transducción de Señal , Moduladores de Tubulina/farmacología , Útero/efectos de los fármacos , Útero/metabolismo , Proteína Wnt4/genética
4.
Biol Reprod ; 99(5): 982-999, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29901777

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Implantación del Embrión/genética , Implantación del Embrión/fisiología , Útero/metabolismo , beta Catenina/metabolismo , Animales , Blastocisto , Línea Celular Tumoral , Estradiol/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos BALB C , Embarazo , Seudoembarazo/genética , Seudoembarazo/metabolismo
5.
Nanomedicine ; 13(6): 1953-1963, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28457934

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Curcumina/farmacología , Neoplasias Endometriales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Micelas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Regulación hacia Abajo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Inmunoterapia , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Rodaminas/metabolismo , Survivin
6.
Bioconjug Chem ; 27(4): 961-72, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26999669

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Estradiol/metabolismo , Receptores de Estrógenos/metabolismo , Nanomedicina Teranóstica , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Dimerización , Femenino , Humanos , Ligandos , Ratones , Ratones Desnudos , Distribución Tisular
7.
Bioorg Med Chem ; 23(4): 839-48, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25618595

RESUMEN

We report herein the design and synthesis of bioisosteres of spirooxindole (MI-63/219), a small-molecule inhibitors of the MDM2-p53 interaction as anti-breast cancer agents. Compound 5b has been exhibiting significant anti-proliferative activity in nude mice bearing MCF-7 xenograft tumor. The compound 5b was found to act via modulation of MDM2 and p53 expression in breast cancer cells expressing wild type p53. Compound 5b stimulated p53 activation, caused modulation of downstream effectors p21, pRb, and cyclin D1 which regulate cell cycle. Thus, compound triggered G1-S phase cell cycle arrest, which was evident by flow cytometric analysis of treated breast cancer cells. Thus, compound 5b restores the p53 function, which triggers molecular events consistent with cell cycle arrest at G1/S phase.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Indoles/química , Indoles/uso terapéutico , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Mama/efectos de los fármacos , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Diseño de Fármacos , Femenino , Humanos , Indoles/farmacología , Ratones Desnudos , Oxindoles , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Compuestos de Espiro/uso terapéutico , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
8.
Toxicol Appl Pharmacol ; 280(2): 323-34, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25123791

RESUMEN

The predominant estrogen-receptor (ER)-ß signaling in normal prostate is countered by increased ER-α signaling in prostate cancer (CaP), which in association with androgen-receptor (AR) signaling results in pathogenesis of the disease. However CaP treatments mostly target AR signaling which is initially effective but eventually leads to androgen resistance, hence simultaneous targeting of ERs has been proposed. A novel series of molecules were designed with multiple sex-steroid receptor modulating capabilities by coalescing the pharmacophores of known anti-CaP molecules that act via modulation of ER(α/ß) and/or AR, viz. 3,3'diindolylmethane (DIM), mifepristone, toremifene, tamoxifen and raloxifene. N,N-diethyl-4-((2-(4-methoxyphenyl)-1H-indol-3-yl)methyl) aniline (DIMA) was identified as the most promising structure of this new series. DIMA increased annexin-V labelling, cell-cycle arrest and caspase-3 activity, and decreased expression of AR and prostate specific antigen in LNCaP cells, in vitro. Concurrently, DIMA increased ER-ß, p21 and p27 protein levels in LNCaP cells and exhibited ~5 times more selective binding for ER-ß than ER-α, in comparison to raloxifene. DIMA exhibited a dose-dependent ER-ß agonism and ER-α antagonism in classical gene reporter assay and decreased hTERT (catalytic subunit of telomerase) transcript levels in LNCaP at 3.0 µM (P<0.05). DIMA also dose-dependently decreased telomerase enzyme activity in prostate cancer cells. It is thus concluded that DIMA acts as a multi-steroid receptor modulator and effectively inhibits proliferation of prostate cancer cells through ER-ß mediated telomerase inhibition, by countering actions of ER-α and AR. Its unique molecular design can serve as a lead structure for generation of potent agents against endocrine malignancies like the CaP.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Indoles/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/efectos de los fármacos , Receptores de Estrógenos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Telomerasa/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Humanos , Masculino , Neoplasias de la Próstata/patología , Receptores Androgénicos/fisiología , Receptores de Estrógenos/fisiología
10.
Med Res Rev ; 32(1): 166-215, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22183797

RESUMEN

Breast cancer is the most common cancer diagnosed in women and the second most common cause of female cancer-related deaths, with more than one million new cases diagnosed per year throughout the world. With the recent advances in the knowledge of cellular processes and signaling pathways involved in the pathogenesis of breast cancer, the current focus of researchers and clinicians is to develop novel treatment strategies that can be included in the armamentarium against breast cancer. With the failure of endocrine-targeted therapy and the development of resistance to existing chemotherapy, the most explored pathway as next generation target for breast cancer therapy has been the epidermal growth factor receptor (EGFR) (ErbB-1)/herceptin-2 (HER-2) (ErbB-2) pathway. This review focuses on the rationale for targeting members of ErbB receptor family and numerous agents that are in use for inhibiting the pathway. The mechanism of action, preclinical and clinical trial data of the agents that are in use for targeting the EGFR/HER-2 pathway and the current status, thereof, have been discussed in detail. In addition, the future clinical trial promises these agents hold either as monotherapy or as combination therapy with conventional agents or with other antisignaling agents have been pondered, so as to provide better and more efficacious treatment strategies for breast cancer patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante/métodos , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Gefitinib , Humanos , Terapia Molecular Dirigida , Quinazolinas/uso terapéutico , Transducción de Señal , Trastuzumab
11.
Reprod Sci ; 29(5): 1560-1576, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34424529

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario , Fertilización , Peroxiredoxina VI , Proteómica , Animales , Trompas Uterinas , Femenino , Oviductos/metabolismo , Peroxiredoxina VI/metabolismo , Embarazo , Proteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Conejos
12.
Am J Obstet Gynecol ; 205(4): 362.e1-11, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21782150

RESUMEN

OBJECTIVE: The study was undertaken to explore the antiproliferative mechanism of action of 2-[piperidinoethoxyphenyl]-3-[4-hydroxyphenyl]-2H-benzo(b)pyran (K-1) in estradiol-induced rat uterine hyperplasia. STUDY DESIGN: Adult ovariectomized rats received vehicle or estradiol alone (20 µg/kg) or estradiol along with K-1 (100 or 200 µg/kg) for 14 days. Uterine histomorphometric analysis and immunoblotting were performed. Caspase-3 activity and terminal deoxynucleotidyl transferase-mediated nick end-labeling staining were performed to analyze the apoptotic potential of compound. RESULTS: Compound inhibited estradiol-induced uterine weight and histomorphometric changes pertaining to endometrial growth and down-regulated the expression of estrogen response element and activator protein-1 regulated genes and transcription factors. The compound significantly induced apoptosis, interfered with Akt activation, decreased X-linked inhibitor of apoptosis protein expression leading to an increased cleavage of caspase-9, caspase-3, poly(adenosine diphosphate-ribose) polymerase, increased Bax/Bcl2 ratio, and caspase-3 activity. CONCLUSION: K-1 inhibits endometrial proliferation via nonclassical estrogen receptor signaling mechanisms. It interfered with Akt activation and induced apoptosis via the intrinsic pathway and inhibited estradiol-induced hyperplasia formation in rat uterus.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzopiranos/farmacología , Hiperplasia/prevención & control , Piperidinas/farmacología , Útero/efectos de los fármacos , Útero/patología , Animales , Femenino , Ratas , Ratas Sprague-Dawley
13.
Bioorg Med Chem ; 19(18): 5409-19, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21871812

RESUMEN

A new series of 1,3-biarylsulfanyl derivatives (homodibenzyl core motif) have been designed and synthesized as new estrogen receptor ligands by chopping benzothiophene core of raloxifene to engender seco-raloxifene scaffold. All the synthesized compounds were screened for anti-proliferative, anti-osteoporotic, and anti-implantation activity. Compounds (35, 36) having basic amino anti-estrogenic side chain were exhibiting potential anti-proliferative activity in MCF-7, MDA-MB-231 and ishikawa cell lines. Some of the synthesized compounds having homodibenzyl motif (5, 8, 10) have shown moderate anti-osteoporotic activity.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Compuestos de Sulfhidrilo/farmacología , Antineoplásicos/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HEK293 , Humanos , Modelos Moleculares , Conformación Molecular , Estereoisomerismo , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/síntesis química , Compuestos de Sulfhidrilo/química
14.
Bioorg Med Chem ; 17(11): 3847-56, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19423356

RESUMEN

A series of 3,4,6-triaryl-2-pyranones, new class of anti-breast cancer agents, have been synthesized as a structural variants of cyclic triphenylethylenes by replacing the fused benzene ring with pendant phenyl ring to mimic the phenolic A ring of estradiol. Nine of these newly synthesized pyranones exhibited significant anti-proliferative activity in both ER+ve and ER-ve breast cancer cell lines. Four active non-cytotoxic compounds 5c, 5d, 5g and 5h showed specific and selective cytotoxicity and two compounds 5d and 5h induced significant DNA fragmentation in both MCF-7 and MDA-MB-231 cell lines. Based on RBA studies, the molecules probably act in an ER-independent mechanism. The involved pathway was observed as caspase-dependant apoptosis in MCF-7 cells. However, the particular caspases involved and the possible cellular target through which this series of compounds mediate cell death are not known.


Asunto(s)
Antineoplásicos/farmacología , Pironas/química , Pironas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Modelos Moleculares , Estructura Molecular , Piperidinas/química , Pirrolidinas/química
15.
Bioorg Med Chem ; 17(19): 6832-40, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19740667

RESUMEN

A novel class of 2,3,4-triarylbenzopyrans has been synthesized and were evaluated for their selective estrogen receptor modulation activity and as a therapeutic agent for breast cancer. Among the compounds synthesized, compounds 11a and 12c exhibited 73.91% and 69.24% inhibition as estrogen antagonistic activity, respectively. Compound 12a showed the lowest IC(50) at 6.97 microM against MCF-7 and 11f showed the lowest IC(50) value of 5.6 microM against MDA-MB-231 cell line in spite of their low receptor binding affinity implicating these compounds probably act through ER independent mechanism.


Asunto(s)
Benzopiranos/síntesis química , Neoplasias de la Mama/tratamiento farmacológico , Moduladores Selectivos de los Receptores de Estrógeno/síntesis química , Benzopiranos/farmacología , Benzopiranos/uso terapéutico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Antagonistas de Estrógenos , Femenino , Humanos , Concentración 50 Inhibidora , Receptores de Estrógenos/metabolismo
16.
Gen Comp Endocrinol ; 160(2): 124-33, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19027743

RESUMEN

The alpha(V)beta(3) integrin as a marker of endometrial receptivity has been well established in human and other mammalian species; however, its expression is still not known in rats. Our objective was to establish the expression of alpha(V)beta(3) integrin as a marker of endometrial receptivity in rat and to further prove its role in implantation by function-blocking studies in this species. Immunocytochemical, immunohistochemical and flow-cytometric studies were performed in rat endometrial epithelial cells (EEC) to demonstrate the expression of alpha(V)beta(3) integrin during non-receptive, pre-receptive and receptive phases of the uterus. Results revealed positive immunocytochemical staining for alpha(v) and beta(3) subunits on the surface of EEC of days 4 and 5p.c. (post-coitum), but the intensity was higher in cells of day 5p.c. Flow-cytometric study revealed higher level of alpha(V)beta(3) on day 5p.c. as compared to day 4p.c. and non-pregnant animals. Immunohistochemical analysis of uterine tissue also revealed that the alpha(V)beta(3) expression in LE was higher on day 5p.c. morning as compared to that observed on day 4p.c. In addition, the expression of beta(3) subunit was not evident in rats receiving ormeloxifene, an agent known to inhibit the uterine receptivity. Immunoblotting experiments also revealed higher expression of uterine beta(3) on day 5p.c. On day 6, expression of beta(3) was high in implantation sites than on inter-implantation sites. In immature ovariectomized rats, alpha(V)beta(3) was up-regulated by progesterone and by a combination of estrogen and progesterone. The expression of alpha(V)beta(3) was also up-regulated in EEC co-cultured with blastocysts. All the agents used for function-blocking studies showed significant reduction in the number of implantation sites in treated horn as compared to sham control horn. The present study has successfully demonstrated the expression of alpha(V)beta(3) in rat EEC as a marker of endometrial receptivity and showed that this molecule is indispensable for the process of implantation in this species.


Asunto(s)
Implantación del Embrión/fisiología , Endometrio/citología , Células Epiteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Animales , Western Blotting , Endometrio/metabolismo , Femenino , Inmunohistoquímica , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Útero/metabolismo
17.
J Mol Med (Berl) ; 97(4): 509-522, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30729278

RESUMEN

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.


Asunto(s)
Decidua/citología , Endometrio/citología , MicroARNs/genética , Proteína Smad1/genética , Células del Estroma/citología , Animales , Proliferación Celular , Decidua/metabolismo , Regulación hacia Abajo , Implantación del Embrión , Endometrio/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Embarazo , Ratas , Ratas Sprague-Dawley , Células del Estroma/metabolismo , Regulación hacia Arriba
18.
J Nutr Biochem ; 70: 215-226, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31252288

RESUMEN

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.


Asunto(s)
Apoptosis , Endometriosis/tratamiento farmacológico , Flavanonas/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Endometrio/efectos de los fármacos , Femenino , Potencial de la Membrana Mitocondrial , Trasplante de Neoplasias , Óxido Nítrico/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Endocrinol ; 240(3): 417-429, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30667362

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Decidua/metabolismo , Implantación del Embrión , Endometrio/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Adulto , Animales , Moléculas de Adhesión Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Proteínas del Citoesqueleto/genética , Decidua/citología , Endometrio/citología , Femenino , Humanos , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , Embarazo , Interferencia de ARN , Células del Estroma/citología , Útero/citología , Útero/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
20.
Eur J Med Chem ; 43(10): 2149-58, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18155810

RESUMEN

Several regioisomeric tetrazolyl indole derivatives with structurally modified alkyl substituents at the tetracyclic indole nitrogen containing N-ethyl amino tetrazole moiety have been synthesized and screened for their ER binding affinity, agonist (estrogenic), antagonist (antiestrogenic) and anti-implantation activities. N-2 regioisomers were found to be moderately antagonists and one compound showed 100% contraceptive efficacy at 10 mg/kg dose. Molecular docking studies carried out in comparison to estradiol and raloxifene showed different binding modes of the two regioisomers to the binding site.


Asunto(s)
Antagonistas de Estrógenos/síntesis química , Antagonistas de Estrógenos/farmacología , Estrógenos/agonistas , Indoles/síntesis química , Indoles/farmacología , Tetrazoles/química , Animales , Anticonceptivos Poscoito/síntesis química , Anticonceptivos Poscoito/química , Anticonceptivos Poscoito/metabolismo , Anticonceptivos Poscoito/farmacología , Diseño de Fármacos , Antagonistas de Estrógenos/química , Antagonistas de Estrógenos/metabolismo , Femenino , Indoles/química , Indoles/metabolismo , Ligandos , Masculino , Modelos Moleculares , Conformación Molecular , Ratas , Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA