Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Curr Issues Mol Biol ; 46(6): 5488-5510, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38921000

RESUMEN

The PHLDA (pleckstrin homology-like domain family) gene family is popularly known as a potential biomarker for cancer identification, and members of the PHLDA family have become considered potentially viable targets for cancer treatments. The PHLDA gene family consists of PHLDA1, PHLDA2, and PHLDA3. The predictive significance of PHLDA genes in cancer remains unclear. To determine the role of pleckstrin as a prognostic biomarker in human cancers, we conducted a systematic multiomics investigation. Through various survival analyses, pleckstrin expression was evaluated, and their predictive significance in human tumors was discovered using a variety of online platforms. By analyzing the protein-protein interactions, we also chose a collection of well-known functional protein partners for pleckstrin. Investigations were also carried out on the relationship between pleckstrins and other cancers regarding mutations and copy number alterations. The cumulative impact of pleckstrin and their associated genes on various cancers, Gene Ontology (GO), and pathway analyses were used for their evaluation. Thus, the expression profiles of PHLDA family members and their prognosis in various cancers may be revealed by this study. During this multiomics analysis, we found that among the PHLDA family, PHLDA1 may be a therapeutic target for several cancers, including kidney, colon, and brain cancer, while PHLDA2 can be a therapeutic target for cancers of the colon, esophagus, and pancreas. Additionally, PHLDA3 may be a useful therapeutic target for ovarian, renal, and gastric cancer.

2.
Curr Issues Mol Biol ; 46(4): 3328-3341, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38666938

RESUMEN

Kidney cancer has emerged as a major medical problem in recent times. Multiple compounds are used to treat kidney cancer by triggering cancer-causing gene targets. For instance, isoquercitrin (quercetin-3-O-ß-d-glucopyranoside) is frequently present in fruits, vegetables, medicinal herbs, and foods and drinks made from plants. Our previous study predicted using protein-protein interaction (PPI) and molecular docking analysis that the isoquercitrin compound can control kidney cancer and inflammation by triggering potential gene targets of IGF1R, PIK3CA, IL6, and PTGS2. So, the present study is about further in silico and in vitro validation. We performed molecular dynamic (MD) simulation, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, cytotoxicity assay, and RT-PCR and qRT-PCR validation. According to the MD simulation (250 ns), we found that IGF1R, PIK3CA, and PTGS2, except for IL6 gene targets, show stable binding energy with a stable complex with isoquercitrin. We also performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the final targets to determine their regulatory functions and signaling pathways. Furthermore, we checked the cytotoxicity effect of isoquercitrin (IQ) and found that 5 µg/mL and 10 µg/mL doses showed higher cell viability in a normal kidney cell line (HEK 293) and also inversely showed an inhibition of cell growth at 35% and 45%, respectively, in the kidney cancer cell line (A498). Lastly, the RT-PCR and qRT-PCR findings showed a significant decrease in PTGS2, PIK3CA, and IGF1R gene expression, except for IL6 expression, following dose-dependent treatments with IQ. Thus, we can conclude that isoquercitrin inhibits the expression of PTGS2, PIK3CA, and IGF1R gene targets, which in turn controls kidney cancer and inflammation.

3.
Mar Drugs ; 20(8)2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35892941

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer and exhibits a devastating 5-year survival rate. The most recent procedure for the treatment of PDAC is a combination of several conventional chemotherapeutic agents, termed FOLFIRINOX, that includes irinotecan, leucovorin, oxaliplatin, and 5-fluorouracil (5-FU). However, ongoing treatment using these agents is challenging due to their severe side effects and limitations on the range of patients available for PDAC. Therefore, safer and more innovative anticancer agents must be developed. The anticarcinoma activity of matairesinol that can be extracted from seagrass has been reported in various types of cancer, including prostate, breast, cervical, and pancreatic cancer. However, the molecular mechanism of effective anticancer activity of matairesinol against pancreatic cancer remains unclear. In the present study, we confirmed the inhibition of cell proliferation and progression induced by matairesinol in representative human pancreatic cancer cell lines (MIA PaCa-2 and PANC-1). Additionally, matairesinol triggers apoptosis and causes mitochondrial impairment as evidenced by the depolarization of the mitochondrial membrane, disruption of calcium, and suppression of cell migration and related intracellular signaling pathways. Finally, matairesinol exerts a synergistic effect with 5-FU, a standard anticancer agent for PDAC. These results demonstrate the therapeutic potential of matairesinol in the treatment of PDAC.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Fluorouracilo/farmacología , Furanos , Humanos , Lignanos , Masculino , Mitocondrias , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
4.
J Cell Physiol ; 236(2): 1025-1042, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32697363

RESUMEN

Osthole is a natural coumarin found in a variety of plants and has been reported to have diverse biological functions, including antimicrobial, antiviral, immunomodulatory, and anticancer effects. Here, we investigated the natural derivative osthole as a promising anticancer compound against ovarian cancer and evaluated its ability to suppress and abrogate tumor progression. In addition, we found the endoplasmic reticulum-mitochondrial axis-mediated anticancer mechanisms of osthole against ES2 and OV90 ovarian cancer cells and demonstrated its calcium-dependent pharmacological potential. Mechanistically, osthole was found to target the phosphatidylinositol 3-kinase/mitogen-activated protein kinase signaling pathway to facilitate tumor suppression in ovarian cancer. Furthermore, we identified the effects of osthole in a three-dimensional tumor-formation model using the zebrafish xenograft assay, providing convincing evidence of the pharmacological effects of osthole within the anchorage-independent tumor microenvironment. These findings suggest that osthole has strong potential as a pharmacological agent for targeting ovarian cancer.


Asunto(s)
Cumarinas/farmacología , Mitocondrias/genética , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Femenino , Humanos , Mitocondrias/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasa/genética , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/genética
5.
Fish Shellfish Immunol ; 112: 135-142, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33746059

RESUMEN

CXCL14 is a chemokine which is orthologous in mammals and fish. CXCL14 has a functional role in different organs, with immunomodulatory functions in mammals, but its expression and function in fish is not well known. Moreover, it shows no effects related to immunity in the central nervous system or the reproductive tract in diverse species. Black rockfish (Sebastes schlegelii) is an economically important fish in Asian countries, whose CXCL14 expression pattern is yet to be understood. In this study, the homology of the CXCL14 amino acid sequence in S. schlegelii was compared with that in other species, including fish. Moreover, in situ hybridization analysis revealed that it was highly expressed in the brain and ovary of S. schlegelii. Taken together, we identified for the first time, the cell-specific expression of CXCL14 in S. schlegelii.


Asunto(s)
Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Peces/genética , Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Quimiocinas CXC/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Filogenia , Alineación de Secuencia/veterinaria
6.
Pestic Biochem Physiol ; 171: 104733, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33357555

RESUMEN

Recently, infertility has become a major global issue. It is crucial to identify environmental factors that lead to infertility. The prevalent use of pesticides in agriculture results in the exposure of livestock and humans to these pesticides. Studies have reported the harmful effects of pesticides on pregnancy. Pyridaben, a pesticide that inhibits mitochondrial complex 1, has been reported to have detrimental effects on neurons, spermatogenesis, hormonal balance, and embryonic development. However, the effect of pyridaben on the female reproductive system has not yet been studied. Therefore, in this study, we evaluated the effects of pyridaben on early pregnancy in porcine reproductive cell lines, which are known to mimic the female reproductive system. Results demonstrated that pyridaben decreased cell growth in porcine endometrial luminal epithelial and porcine trophectoderm cell lines through inhibition of cell signal transduction. Further, pyridaben increased subG1 phase and late apoptosis through the induction of reactive oxygen species production, mitochondrial dysfunction, calcium unbalances, pro-apoptotic signals, and endoplasmic reticulum (ER) stress. Moreover, we found that pyridaben induced autophagy and inhibition of placentation through the regulation of ER-mitochondria axis proteins. Overall, pyridaben was found to be harmful in early pregnancy in pigs and may have similar effects in human pregnancy.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Animales , Muerte Celular , Proliferación Celular , Femenino , Embarazo , Piridazinas , Especies Reactivas de Oxígeno , Porcinos
7.
Pestic Biochem Physiol ; 179: 104974, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34802524

RESUMEN

Folpet, a fungicide, is utilized even in cosmetics and pharmaceuticals. The LD50 of folpet in mammals, birds, and fish is relatively high. Recently, several negative effects of folpet on the respiratory system and cornea have been reported. However, there is no study on the negative effects of folpet on maternal-fetus interactions. In the present study, we used porcine trophectoderm (pTr) cells and porcine luminal epithelial (pLE) cells to investigate the toxic effects of folpet during implantation. Folpet treatment decreased cell proliferation and promoted apoptosis with cell cycle arrest. In addition, the ERK, JNK, and AKT signal pathways were activated by folpet treatment. Folpet treatment induced calcium overload in pTr and pLE cells mediating antimigratory and antiadhesive effects in both cell lines. Co-treatment with calcium chelates decreased the anti-implantation effect of folpet. Overall, our results demonstrated potential reproductive toxicity of folpet in pig.


Asunto(s)
Calcio , Fosfatidilinositol 3-Quinasas , Animales , Células Epiteliales , Homeostasis , Ftalimidas , Embarazo , Porcinos
8.
J Pineal Res ; 69(2): e12670, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32421880

RESUMEN

Melatonin has been shown to improve in vitro fertilization and offspring survival after bacterial infection, but its role in regulating maternal-fetal communication during early pregnancy has not been investigated. Results of this study demonstrated expression of abundant melatonin receptors in conceptus and endometrium during early pregnancy. In gilts, expression of melatonin receptor 1A (MTNR1A or MT1) and melatonin receptor 1B (MTNR1B or MT2) increased in trophectoderm (Tr) and uterine luminal epithelium (LE) with advancing days during early pregnancy in a different manner. Melatonin increased proliferation and migration of porcine trophectoderm (pTr) cell, the percent pTr cells in the G2 phase of the cell cycle, and the expression of implantation-related genes by pTr cells and endometrial luminal epithelium (pLE). Melatonin also attenuated the production of LPS-induced pro-inflammatory cytokines and tunicamycin-induced endoplasmic reticulum (ER) stress-sensing proteins. The expression of sirtuin 1 (SIRT1) as a potential target of melatonin increased between Days 9 and 14 of gestation. Co-treatment with SIRT1 inhibitor EX527 and melatonin restored cell-cell interactions through PI3K and MAPK signaling. Knockdown of SIRT1 decreased the expression of implantation-related genes, as well as migration of pTr and pLE cells. The expression of microRNAs regulated by SIRT1 was suppressed in response to melatonin. Furthermore, melatonin significantly increased lipopolysaccharide (LPS)-reduced fertilization and embryogenesis in zebrafish model. These results suggest that melatonin may improve the uterine-conceptus interactions via the regulation of SIRT1 during early pregnancy.


Asunto(s)
Embrión de Mamíferos/embriología , Intercambio Materno-Fetal/efectos de los fármacos , Melatonina/farmacología , Sirtuina 1/metabolismo , Útero/metabolismo , Animales , Femenino , Embarazo , Porcinos
9.
Mar Drugs ; 18(5)2020 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-32429354

RESUMEN

Ovarian cancer is difficult to diagnose early and has high rates of relapse and mortality. Therefore, the treatment of ovarian cancer needs to be improved. Recently, several studies have been conducted in an attempt to develop anticancer drugs from naturally derived ingredients. Compared to traditional chemotherapy, natural compounds can overcome drug resistance with lower side effects. Fucosterol, a phytosterol present in brown algae, reportedly possesses many bioactive effects, including anticancer properties. However, the anticancer effects of fucosterol in ovarian cancer remain unexplored. Therefore, we investigated the effects of fucosterol on progression in human ovarian cancer cells. Fucosterol inhibited cell proliferation and cell-cycle progression in ovarian cancer cells. Additionally, fucosterol regulated the proliferation-related signaling pathways, the production of reactive oxygen species, mitochondrial function, endoplasmic reticulum stress, angiogenesis, and calcium homeostasis. Moreover, it decreased tumor formation in a zebrafish xenograft model. These results indicate that fucosterol could be used as a potential therapeutic agent in ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Phaeophyceae , Estigmasterol/análogos & derivados , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Océanos y Mares , Neoplasias Ováricas/tratamiento farmacológico , Estigmasterol/farmacología , Estigmasterol/uso terapéutico , Pez Cebra
10.
Mar Drugs ; 18(1)2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936539

RESUMEN

Marine organisms are sources of several natural compounds with potential clinical use. However, only a few marine-based pharmaceuticals have been approved for use due to limited knowledge on their biological activities. Here, we identified the functional role of fucoidan extracted from Fucus vesiculosus on ovarian cancer. Fucoidan increased the death of ES-2 and OV-90 cells, through a reduction in proliferation, cell cycle arrest, releases of cytochrome c, reactive oxygen species (ROS) generation, and endoplasmic reticulum (ER) stress. Additionally, fucoidan increased the concentration of cytosolic and mitochondrial calcium in both cells. The decrease of cell proliferation was controlled by the inactivation of PI3K and MAPK signaling cascades in ES-2 and OV-90 cells. In a toxicity assay with normal zebrafish larvae, fucoidan did not induce toxicity, cardiotoxicity, development, kinesis, and apoptosis at different concentrations. However, it disrupted tumor formation and vascular development in a zebrafish xenograft model and angiogenesis transgenic (Tg, fli1-eGFP) model, respectively. Collectively, the results indicate that fucoidan may be a novel pharmaceutical for the management of human ovarian cancer.


Asunto(s)
Calcio/metabolismo , Fucus/química , Homeostasis/efectos de los fármacos , Polisacáridos/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Femenino , Humanos , Neovascularización Patológica/prevención & control , Neoplasias Ováricas/tratamiento farmacológico , Polisacáridos/toxicidad , Pruebas de Toxicidad , Pez Cebra
11.
Mar Drugs ; 18(3)2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182828

RESUMEN

Ovarian cancer (OC) is difficult to diagnose at an early stage and leads to the high mortality rate reported in the United States. Standard treatment for OC includes maximal cytoreductive surgery followed by platinum-based chemotherapy. However, relapse due to chemoresistance is common in advanced OC patients. Therefore, it is necessary to develop new anticancer drugs to suppress OC progression. Recently, the anticancer effects of laminarin, a beta-1,3-glucan derived from brown algae, have been reported in hepatocellular carcinoma, colon cancer, leukemia, and melanoma. However, its effects in OC are not reported. We confirmed that laminarin decreases cell growth and cell cycle progression of OC cells through the regulation of intracellular signaling. Moreover, laminarin induced cell death through DNA fragmentation, reactive oxygen species generation, induction of apoptotic signals and endoplasmic reticulum (ER) stress, regulation of calcium levels, and alteration of the ER-mitochondria axis. Laminarin was not cytotoxic in a zebrafish model, while in a zebrafish xenograft model, it inhibited OC cell growth. These results suggest that laminarin may be successfully used as a novel OC suppressor.


Asunto(s)
Antineoplásicos/uso terapéutico , Línea Celular Tumoral/efectos de los fármacos , Glucanos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Phaeophyceae , Antineoplásicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Glucanos/farmacología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fitoterapia
12.
Dev Biol ; 441(1): 146-158, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30056935

RESUMEN

Chemokines and their receptors play a crucial role in embryo implantation at the maternal-fetal interface during pregnancy. In this study, we investigated the role of CCL4 in development of the porcine endometrium in the early gestational period. Porcine CCL4 showed high similarity with the human counterpart, and mRNA expression of CCL4 and its receptor (CCR5) was predominantly present in the endometrium during early pregnancy. Treatment with CCL4 increased proliferation of porcine uterine luminal epithelial (pLE) cells by activation of PI3K and MAPK signal transduction. In addition, CCL4 recovered the endoplasmic-reticulum stress-reduced proliferation and decreased the unfolded protein response in pLE cells. Besides, the lipopolysaccharide-activated NF-κB pathway was suppressed in response to CCL4 in pLE cells. Inhibition of CCR5 decreased the proliferation of pLE cells and activation of the PI3K and MAPK pathways by CCL4. Furthermore, CCL4 enhanced conceptus-maternal interactions between porcine trophectoderm (pTr) cells and pLE cells during early pregnancy by activating expression of migration and implantation-related genes. Collectively, the results suggest that CCL4 may improve successful implantation in early pregnancy in pigs.


Asunto(s)
Quimiocina CCL4/metabolismo , Implantación del Embrión/fisiología , Endometrio/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Placenta/metabolismo , Embarazo/metabolismo , Porcinos/embriología , Animales , Proliferación Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores CCR5/metabolismo
13.
J Cell Physiol ; 234(4): 4864-4873, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30238980

RESUMEN

Ephrin A1 has a role in a variety of biological events, including cell proliferation, differentiation, migration, and angiogenesis. Ephrin A1 expression is abundant in trophoblasts and endometrial cells during the implantation period; however, its intracellular activities have not yet been reported in bovine endometrial (BEND) epithelial cells. The aim of this study was to identify the functional role of ephrin A1 in BEND cells, which have served as a good model system for investigating the regulation of signal transduction following treatment with interferon-τ (IFNT) in vitro. Supplementation of ephrin A1 to BEND cells increased cell proliferation and increased levels of proliferating cell nuclear antigen and cyclin D1 protein in BEND cell nuclei. To investigate intracellular mechanisms regulated by ephrin A1, we performed Western blot analysis focused on mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling, which are significantly involved in the successful maintenance of pregnancy. Ephrin A1 dose-dependently increased phosphorylation of extracellular signal-regulated kinases (ERK)1/2, c-Jun N-terminal kinases (JNK), P38, protein kinase B (AKT), P70S6K, S6, and cyclin D1, and the activated proteins were suppressed by pharmacological inhibitors including wortmannin (a PI3K inhibitor), U0126 (an ERK1/2 inhibitor), and SP600125 (a JNK inhibitor). Among ephrin A1 receptors, abundant expression of EPHA2 and EPHA4 messenger RNA was detected in BEND cells by reverse transcription polymerase chain reaction analysis. Furthermore, tunicamycin-induced endoplasmic reticulum (ER) stress was inactivated by ephrin A1 treatment of BEND cells. Our findings suggest that ephrin A1 promotes the development of BEND cells and likely enhances uterine capacity and maintenance of pregnancy by activating MAPK and PI3K signaling cascades and by restoring ER stress.


Asunto(s)
Ciclina D1/metabolismo , Endometrio/metabolismo , Efrina-A1/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Antracenos/farmacología , Butadienos/farmacología , Bovinos , Ciclo Celular/fisiología , Línea Celular , Proliferación Celular , Endometrio/citología , Estrés del Retículo Endoplásmico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Interferón Tipo I/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Embarazo , Proteínas Gestacionales/farmacología , Receptor EphA2/metabolismo , Receptor EphA4/metabolismo , Wortmanina/farmacología
14.
J Cell Physiol ; 233(4): 3295-3305, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28885691

RESUMEN

Fibroblast growth factor 2 (FGF2) is abundantly expressed in conceptuses and endometria during pregnancy in diverse animal models including domestic animals. However, its intracellular mechanism of action has not been reported for bovine endometrial cells. Therefore, the aim of this study was to identify functional roles of FGF2 in bovine endometrial (BEND) cell line which has served as a good model system for investigating regulation of signal transduction following treatment with interferon-tau (IFNT) in vitro. Results of present study demonstrated that administration of FGF2 to BEND cells increased their proliferation and regulated the cell cycle through DNA replication by an increase of PCNA and Cyclin D1. FGF2 also increased phosphorylation of AKT, P70S6K, S6, ERK1/2, JNK, and P38 in BEND cells in a dose-dependent manner, and expression of each of those transcription factors was inhibited by their respective pharmacological inhibitor including Wormannin, U0126, and SP600125. In addition, the increase in proliferation of BEND cells and activation of the protein kinases in response to FGF2 was suppressed by BGJ398, a FGFR inhibitor. Furthermore, proliferation of BEND cells was inhibited by tunicamycin, but treatment of BEND cells with FGF2 restored proliferation of BEND cells. Consistent with this result, the stimulated unfolded protein response (UPR) regulatory proteins induced by tunicamycin were down-regulated by FGF2. Results of this study suggest that FGF2 promotes proliferation of BEND cells and likely enhances uterine capacity and maintenance of pregnancy by activating cell signaling via the PI3K and MAPK pathways and by restoring ER stress through the FGFR.


Asunto(s)
Endometrio/citología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fase G2/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitosis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Bovinos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Modelos Biológicos , Fosforilación/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
15.
J Cell Physiol ; 233(4): 3529-3539, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29023770

RESUMEN

To reduce embryonic losses in domestic animals for economic production of livestock meat and milk, chemokines and their receptors are required for proper implantation and placentation during early pregnancy. In this study, we investigated the effects of C-C-motif chemokine ligand 23 (CCL23) on the proliferation of bovine endometrial (BEND) epithelial cells. CCL23 treatment improved BEND cell proliferation by enhancing PCNA and cyclin D1 expression via activation of the PI3K/AKT and MAPK signaling pathways. In addition, a combination of CCL23 and tunicamycin reversed the ER stress-induced reduction in cell proliferation and the decreased expression of UPR-mediated signaling proteins, including IRE1α, PERK, and ATF6α. Moreover, it regulated the lipopolysaccharide-induced inflammation in BEND cells by inhibiting the expression of pro-inflammatory cytokines (IL-6 and IL-8), and by restoring intracellular Ca2+ levels. These findings demonstrate that CCL23 improves endometrial development and uterine receptivity required for implantation and placentation during early pregnancy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Endometrio/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Animales , Bovinos , Implantación del Embrión/efectos de los fármacos , Endometrio/metabolismo , Células Epiteliales/metabolismo , Femenino , Lipopolisacáridos/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos
16.
J Cell Physiol ; 233(4): 3141-3151, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28816360

RESUMEN

Chemokines play an important role in regulating the complex immune system at the maternal-fetal interface during pregnancy. Among various chemokines, CC motif chemokine ligand 2 (CCL2) plays a role in the recruitment of immune regulatory cells to implantation sites within the endometrium. In cattle, CCL2 is abundantly expressed in the uterine endometrium. However, its intracellular signaling has not been identified. In this study, we examined the effects of CCL2 on bovine endometrial (BEND) cell proliferation. CCL2 stimulated BEND cell proliferation by abundant expression of PCNA, accumulation of cells in the G2/M phase, and activation of the PI3 K/AKT and MAPK signaling pathways. Moreover, CCL2 reduced endoplasmic reticulum stress and restored the inflammation-induced reduction in BEND cell proliferation by regulating the unfolded protein response genes and cytokines. Collectively, these results demonstrated that CCL2 plays a pivotal role in reproductive tissues and may support maternal-fetal interface to improve efficiency of pregnancy.


Asunto(s)
Endometrio/patología , Estrés del Retículo Endoplásmico , Células Epiteliales/patología , Inflamación/patología , Lipopolisacáridos/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/química , Quimiocina CCL2/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Secuencia de ADN , Tunicamicina/farmacología
17.
J Cell Physiol ; 233(3): 2560-2571, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28777434

RESUMEN

Structural and functional development of the mammary gland is constant in the mammary gland life cycle. Eph receptors and their ligands, ephrins, control events through cell-to-cell interactions during embryonic development, and adult tissue homeostasis; however, little information on participation of ephrin A1, a representative ligand of the Eph receptor, in the development and function of normal mammary glands is known. In this study, we demonstrated functional effects of the ephrin A1-Eph system and mechanisms of its action on bovine mammary epithelial (MAC-T) cells. The in vitro cultured MAC-T cells expressed the ephrin A1 ligand and EphA1, A2, A4, A7, and A8 among the eight members of the Eph A family. Our results revealed that ephrin A1 induced MAC-T cell cycle progression and stimulated cell proliferation with abundant expression of nucleic PCNA and cyclin D1 proteins. Additionally, ephrin A1 induced activation of intracellular signaling molecules involved in PI3 K/AKT and MAPK signaling, and the proliferation-stimulating effect of ephrin A1 was mediated by activation of these pathways. Furthermore, ephrin A1 influenced expression and activation of various ER stress-related proteins and protected MAC-T cells from stress-induced cell death. Finally, ephrin A1 alleviated LPS-induced cell death through down-regulation of inflammatory cytokines. In conclusion, the results of this study suggest that the Eph A-ephrin A1 system is a positive factor in the increase and maintenance of epithelial cells in mammary glands of cows; the signaling system contributes to development, remodeling, and functionality of normal mammary glands and could overcome mastitis in cows and other mammals.


Asunto(s)
Proliferación Celular , Estrés del Retículo Endoplásmico , Efrina-A1/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Mastitis/metabolismo , Receptor EphA1/metabolismo , Animales , Bovinos , Muerte Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Estrés del Retículo Endoplásmico/efectos de los fármacos , Efrina-A1/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Lipopolisacáridos/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Mastitis/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphA1/efectos de los fármacos , Transducción de Señal
18.
J Dairy Sci ; 101(5): 4527-4541, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29454702

RESUMEN

C-C motif chemokine ligand 2 (CCL2) is a small chemokine which belongs to the CC-type chemokine family, and has chemoattractant activity for recruitment of monocytes to sites of inflammation. Overexpressed CCL2 binding to its receptor C-C chemokine receptor 2 increases the risk of breast cancer in humans, but its effects on proliferation of bovine mammary epithelial cells is not known. Maintaining a high level of proliferative activity in bovine mammary epithelial cells during lactation is important for improving milk yield and can benefit the dairy industry economically. In the present study, we demonstrated that CCL2 induces proliferation of MAC-T cells, a bovine mammary epithelial cell line, and stimulates progression of the cell cycle through stimulation of expression of cyclin D1. Moreover, CCL2 activates phosphoinositide 3-kinase (PI3K)/AKT [AKT, P70-S6 kinase 1 (P70S6K), ribosomal protein S6 (S6)] and mitogen activated protein kinase (MAPK) [extracellular signal-regulated kinase-1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and P38] pathways, which are involved in proliferation of MAC-T cells, as evidenced by co-treatment of MAC-T cells with pharmacological inhibitors of cell signaling transcription factors including Wortmannin, U0126, and SP600125. The CCL2 in MAC-T cells attenuates endoplasmic reticulum stress induced by tunicamycin, suggesting that CCL2 regulates intracellular synthesis of proteins and lipids and prevents activation of apoptotic pathways initiated in response to endoplasmic reticulum stress. Furthermore, CCL2 is involved in alleviating lipopolysaccharide (LPS)-induced inflammatory responses in MAC-T cells by reducing LPS-induced expression of IL8, IL6, and nuclear factor kappa B subunit 1 (NFKB1). Collectively, CCL2 is a novel target for improving the quantity and quality of milk from cows through stimulation of proliferation on mammary epithelial cells and attenuation of LPS-induced inflammatory responses.


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Proliferación Celular/efectos de los fármacos , Inflamación/veterinaria , Lipopolisacáridos/farmacología , Glándulas Mamarias Animales/patología , Receptores CCR2/fisiología , Animales , Bovinos , Enfermedades de los Bovinos/patología , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/fisiología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Inflamación/inducido químicamente , Inflamación/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lactancia/efectos de los fármacos , Leche/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos
19.
J Cell Physiol ; 232(6): 1527-1538, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27775163

RESUMEN

Eph and ephrin regulate diverse biological events such as proliferation, adhesion, migration, and angiogenesis through cell-to-cell interactions. However, little is known of their functional role and mechanisms of action in uterine endometrial cells. In the present study, we demonstrated the effects of the Eph and ephrin on interactions between blastocysts and endometrial luminal epithelial (pLE) cells in the pig that is regarded as an excellent biomedical animal model for research on the peri-implantation period of pregnancy. Results of this study indicated that among eight members of the Eph A family, expression of Eph A1, A2, A4, and A7 was strongly detected in endometrial epithelial cells during early pregnancy. Of these, for identification of signal transduction pathways induced by ephrin A1, a major ligand for Eph A, cell proliferation assays, and immunofluorescence and cell cycle regulation were analyzed following treatment of pLE cells with ephrin A1. Ephrin A1 stimulated proliferation of pLE cells as evidenced by abundant PCNA expression and an increase in the G2/M phase. Western blot analysis showed that ephrin A1 activated PI3K and MAPK signaling proteins in a time-dependent manner. Moreover, phosphorylation of AKT, ERK1/2, P38, and JNK proteins were suppressed by their inhibitors wortmannin, U0126, SB203580, and SP600125, respectively. Also, phosphor-AKT was reduced by ERK1/2 and P38 inhibitors. Ephrin A1-induced proliferation and migration of pLE cells was also blocked by those inhibitors. Collectively, these results suggest that ephrin A1 enhances interactions between porcine blastocysts and endometrial luminal epithelial cells by activating PI3K and MAPK signal transduction pathways. J. Cell. Physiol. 232: 1527-1538, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Endometrio/citología , Efrina-A1/metabolismo , Células Epiteliales/metabolismo , Receptores de la Familia Eph/metabolismo , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Ciclo Estral , Femenino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Embarazo , Transducción de Señal , Sus scrofa , Factores de Tiempo
20.
J Cell Physiol ; 232(12): 3641-3651, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28150859

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family binds to two transmembrane receptors; neurotrophic receptor tyrosine kinase 2 (NTRK2) with high affinity and p75 with low affinity. Although BDNF-NTRK2 signaling in the central nervous system is known, signaling in the female reproductive system is unknown. Therefore, we determined effects of BDNF on porcine endometrial luminal epithelial (pLE) cells isolated from Day 12 of pregnancy, as well as expression of BDNF and NTRK2 in endometria of cyclic and pregnant pigs. BDNF-NTRK2 genes were expressed in uterine glandular (GE) and luminal (LE) epithelia during early pregnancy. In addition, their expression in uterine GE and LE decreased with increasing parity of sows. Recombinant BDNF increased proliferation in pLE cells in a dose-dependent, as well as expression of PCNA and Cyclin D1 in nuclei of pLE cells. BDNF also activated phosphorylation of AKT, P70S6K, S6, ERK1/2, JNK, P38 proteins in pLE cells. In addition, cell death resulting from tunicamycin-induced ER stress was prevented when pLE cells were treated with the combination of tunicamycin and BDNF which also decreased cells in the Sub-G1 phase of the cell cycle. Furthermore, tunicamycin-induced unfolded protein response genes were mostly down-regulated to the basal levels as compared to non-treated pLE cells. Our finding suggests that BDNF acts via NTRK2 to induce development of pLE cells for maintenance of implantation and pregnancy by activating cell signaling via the PI3K and MAPK pathways and by inhibiting ER stress.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Proliferación Celular/efectos de los fármacos , Endometrio/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Endometrio/citología , Endometrio/metabolismo , Células Epiteliales/metabolismo , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Edad Gestacional , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Paridad , Fosforilación , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkB/agonistas , Receptor trkB/genética , Receptor trkB/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factores de Tiempo , Tunicamicina/toxicidad , Respuesta de Proteína Desplegada/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA