Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Theriogenology ; 226: 39-48, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38838613

RESUMO

Prokineticin 1 (PROK1) is an important factor in pregnancy establishment in pigs, acting at the embryo-maternal interface and the corpus luteum (CL). Estradiol-17ß (E2) is the primary pregnancy recognition signal in pigs, and its effects are augmented by luteotropic prostaglandin E2 (PGE2). On the contrary, prostaglandin F2α (PGF2α) exerts mainly a luteolytic effect. The present study aimed to elucidate whether E2, PGE2, and PGF2α regulate the expression of PROK1 and its receptors in the porcine CL and to determine the PROK1 effect on luteal endothelial cells and pathways that may be involved in this regulation. The effects of E2, PGE2, and PGF2α on the expressions of PROK1 and its receptors in the CL were studied using an in vitro model of ultrathin luteal tissue explants model. Additionally, the effects of E2 and PGE2 on the PROK1 system were determined using an in vivo approach, in which the hormones were administered into the uterine lumen to imitate their secretion by embryos. Endothelial cell proliferation was measured using the colorimetric method. E2 acting via estrogen receptors simulated the mRNA and protein expressions of PROK1 and PROKR1 in CL explants in vitro (p < 0.05). The simultaneous action of E2 with PGE2 enhanced the expression of luteal PROK1 mRNA in vitro (p < 0.05). Estradiol-17ß acting alone significantly increased PROK1 mRNA levels in vivo, whereas E2 simultaneously administered with PGE2 significantly elevated the PROK1 mRNA expression and PROKR1 mRNA and protein contents in CLs adjacent to uterine horns receiving hormonal infusion compared with CLs adjacent to placebo-treated uterine horns (p < 0.01). The PROK1 protein expression was significantly higher in the CLs of pigs treated with E2, PGE2, and E2 together with PGE2 than in the control group. PGF2α increased the PROK1 mRNA content in CLs on days 12 and 14 of the estrous cycle (p < 0.05). The expression of PROKR2 at the mRNA and protein levels remained unchanged in response to in vitro and in vivo treatments. PROK1 stimulated the proliferation of luteal endothelial cells by activating the MAPK, AKT, and mTOR pathways (p < 0.05). In summary, the luteal expressions of PROK1 and PROKR1 in early pregnancy are regulated by E2 and PGE2. PROK1 stimulates luteal angiogenesis by activating the MAPK, AKT, and mTOR pathways. The regulation of luteal PROK1 expression by PGF2α indicates PROK1's putative role during luteolysis. We conclude that PROK1-PROKR1 signaling supports luteal function during CL rescue in pregnancy in pigs.


Assuntos
Corpo Lúteo , Hormônios Gastrointestinais , Regulação da Expressão Gênica , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Animais , Feminino , Gravidez , Corpo Lúteo/metabolismo , Corpo Lúteo/efeitos dos fármacos , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Estradiol/farmacologia , Estradiol/metabolismo , Hormônios Gastrointestinais/metabolismo , Hormônios Gastrointestinais/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Suínos , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
2.
Int J Med Sci ; 21(1): 27-36, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38164347

RESUMO

Prokineticin 1 (PROK1) is a secreted protein involved in a range of physiological activities such as cell proliferation, migration, angiogenesis, and neuronal cell proliferation. Emerging evidences show that PROK1/PROK receptors (PROKRs) are expressed by trophoblasts, and decidual stroma cells at the maternal-fetal interface. PROK1 plays a critical role in successful pregnancy establishment by regulating the decidualization, implantation and placental development. Dysregulation of prokineticin signaling has been described in certain pathological states associated with pregnancy, including pre-eclampsia, recurrent miscarriage and fetal growth restriction. In this review, the expression and pleiotropic roles of PROK1 under physiological and pathological pregnancy conditions are discussed.


Assuntos
Hormônios Gastrointestinais , Pré-Eclâmpsia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Gravidez , Feminino , Humanos , Placenta/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Transdução de Sinais/genética , Trofoblastos , Pré-Eclâmpsia/genética , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo
3.
Sci Rep ; 13(1): 5085, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36991037

RESUMO

Prokineticin 1 (PROK1) is a pleiotropic factor secreted by endocrine glands; however, its role has not been studied in the corpus luteum (CL) during pregnancy in any species. The present study aimed to investigate the contribution of PROK1 in regulating processes related to porcine CL function and regression: steroidogenesis, luteal cell apoptosis and viability, and angiogenesis. The luteal expression of PROK1 was greater on Days 12 and 14 of pregnancy compared to Day 9. PROK1 protein expression during pregnancy increased gradually and peaked on Day 14, when it was also significantly higher than that on Day 14 of the estrous cycle. Prokineticin receptor 1 (PROKR1) mRNA abundance increased on Days 12 and 14 of pregnancy, whereas PROKR2 elevated on Day 14 of the estrous cycle. PROK1, acting via PROKR1, stimulated the expression of genes involved in progesterone synthesis, as well as progesterone secretion by luteal tissue. PROK1-PROKR1 signaling reduced apoptosis and increased the viability of luteal cells. PROK1 acting through PROKR1 stimulated angiogenesis by increasing capillary-like structure formation by luteal endothelial cells and elevating angiogenin gene expression and VEGFA secretion by luteal tissue. Our results indicate that PROK1 regulates processes vital for maintaining luteal function during early pregnancy and the mid-luteal phase.


Assuntos
Células Lúteas , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Gravidez , Feminino , Animais , Suínos , Progesterona/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Células Endoteliais/metabolismo , Corpo Lúteo/metabolismo , Células Lúteas/metabolismo
4.
Biomed Res Int ; 2022: 2309339, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845958

RESUMO

Lower-grade glioma (LGG) is a crucial pathological type of glioma. Prokineticins have not been reported in LGG. Prokineticins as a member of the multifunctional chemokine-like peptide family are divided into two ligands: PROK1 and PROK2. We evaluated the role of PROK1 and PROK2 in LGG using TCGA database. We downloaded the datasets of LGG from TCGA and evaluated the influence of prokineticins on LGG survival by survival module. Correlations between clinical information and prokineticins expression were analyzed using logistic regression. Univariable survival and multivariate Cox analysis was used to compare several clinical characteristics with survival. Correlation between prokineticins and cancer immune infiltrates was explored using CIBERSORT and correlation module of GEPIA. We analyzed genes of PROK1 and PROK2 affecting LGG, screened differentially expressed genes (DEGs), interacted protein-protein with DEGs through the STRING website, then imported the results into the Cytospace software, and calculated the hub genes. To analyze whether hub genes and prokineticins are related, the relationship between PROK1 and PROK2 and hub genes was assessed and shown by heat map. In addition, gene set enrichment analysis (GSEA) was performed using the TCGA dataset. The univariate analysis using logistic regression and PROK1 and PROK2 showed opposite expression differences between tumor and normal tissues (p < 0.05). PRO1 and PROK2 expressions showed significant differences in tumor grade, age, Iiscitrate DeHydrogenase (IDH) status, histological type, and 1P/19q codeletion. Multivariate analysis revealed that the up-regulated PROK1 and PROK2 expression is an independent prognostic factor for bad prognosis. Specifically, prokineticin expression level has significant correlations with infiltrating levels of Th1 cells, NK CD 56bright cells, and Mast cells in LGG. We screened 21 DEGs and obtained 5 hub genes (HOXC10, HOXD13, SOX4, GATA4, HOXA9). GSEA-identified FCMR activation, creation of C4 and C2 activators, and CD22-mediated BCR regulation in gene ontology (GO) were differentially enriched in high PROK1 and PROK2 expression phenotype pathway, cytoplasmic ribosomal proteins, and ribosome and were differentially enriched in the low PROK1 and PROK2 expression phenotype pathway. Prokineticins are a prognostic biomarker and the correlation between hub genes and LGG requires further attention.


Assuntos
Neoplasias Encefálicas , Hormônios Gastrointestinais , Glioma , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina , Biomarcadores , Neoplasias Encefálicas/patologia , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Ontologia Genética , Glioma/patologia , Proteínas de Homeodomínio , Humanos , Prognóstico , Fatores de Transcrição SOXC , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
5.
Cell Biochem Funct ; 39(2): 308-316, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32876972

RESUMO

Cancer cell derived exosomes play important roles in cancer progression and modulation of the tumour microenvironment. This study aims to investigate the role of prokineticin receptor 1 (PKR1) positive exosomes on angiogenesis. In the present study, PKR1 expression in tumour samples from ovarian cancer patients were examined firstly. Then, two ovarian cancer cell lines, namely A2780 and HO-8910 cells, were used to isolate and obtain the PKR1 positive exosomes from the serum free medium. The function analysis of PKR1 positive exosomes on angiogenesis was conducted by cell proliferation and migration assay, tube formation analysis, and tumour volume assay. The results showed that PKR1 expression was down regulated in tumour samples of ovarian cancer patients compared with adjacent normal tissues. The intracellular expression of PKR1 could be detected in A2780 and HO-8910 cells. And, the isolated exosomes from the serum free medium were confirmed by transmission electron microscopic and NTA analysis, as well as the co-presence of PKR1 with exosome marker CD63. The function analysis of PKR1 positive exosomes on angiogenesis demonstrated the uptake of PKR1 positive exosomes by human umbilical vein endothelial cells through immunofluorescence staining. The angiogenesis assays in vitro indicated that PKR1 positive exosomes promoted migration and tube formation of HUVECs but not proliferation. The endogenous PKR1 was also verified to help to enhance migration and promote tube formation of vascular endothelial cells, which might involved in the phosphorylation of STAT3. Additionally, The tumour volume from exosomes treated A2780 tumour-bearing mice was significantly increased compared with the control group, accompanied with the induced PKR1 expression and phosphorylation of STAT3 level. SIGNIFICANCE OF THE STUDY: This study proved the important role of PKR1 positive exosomes released from ovarian cancer cells on promoting angiogenesis. The data indicated that PKR1 derived from ovarian cancer cells could act as an important tumour associated antigen and biomolecular factor for cellular communication in tumour microenvironment.


Assuntos
Exossomos/metabolismo , Hormônios Gastrointestinais/metabolismo , Neovascularização Fisiológica , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Exossomos/transplante , Feminino , Hormônios Gastrointestinais/antagonistas & inibidores , Hormônios Gastrointestinais/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transplante Heterólogo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/antagonistas & inibidores , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
6.
Pharmacol Res ; 160: 105190, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32937177

RESUMO

Heart and brain development occur simultaneously during the embryogenesis, and both organ development and injuries are interconnected. Early neuronal and cardiac injuries share mutual cellular events, such as angiogenesis and plasticity that could either delay disease progression or, in the long run, result in detrimental health effects. For this reason, the common mechanisms provide a new and previously undervalued window of opportunity for intervention. Because angiogenesis, cardiogenesis and neurogenesis are essential for the development and regeneration of the heart and brain, we discuss therein the role of prokineticin as an angiogenic neuropeptide in heart-brain development and injuries. We focus on the role of prokineticin signaling and the effect of drugs targeting prokineticin receptors in neuroprotection and cardioprotection, with a special emphasis on heart failure, neurodegenerativParkinson's disease and ischemic heart and brain injuries. Indeed, prokineticin triggers common pro-survival signaling pathway in heart and brain. Our review aims at stimulating researchers and clinicians in neurocardiology to focus on the role of prokineticin signaling in the reciprocal interaction between heart and brain. We hope to facilitate the discovery of new treatment strategies, acting in both heart and brain degenerative diseases.


Assuntos
Encefalopatias/genética , Encéfalo/crescimento & desenvolvimento , Cardiopatias/genética , Coração/crescimento & desenvolvimento , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Animais , Encéfalo/fisiologia , Coração/fisiologia , Humanos , Neurogênese/genética , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia
7.
J Gynecol Obstet Hum Reprod ; 49(9): 101835, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32585394

RESUMO

The aim of the present study was to investigate the expression patterns of prokineticins (PROK) and prokineticin receptors (PROKR) in the endometrium of women with recurrent implantation failure (RIF). Fifteen (15) women with RIF and 15 fertile controls were enrolled in this study. Endometrial samples were taken from study participants with an endometrial biopsy cannula during the implantation window. Real time polymerase chain reaction and immunohistochemistry were used to determine PROK/PROKR mRNA expression and protein localization, respectively. PROK1 mRNA levels were 6.09 times higher compared to endometrial samples obtained from women with RIF than in samples obtained from fertile controls, whereas PROKR1 mRNA levels were 2.46 times lower in endometrial samples obtained from women with RIF than in samples from fertile controls. In addition, decreased PROKR1 was supported by immunohistochemistry analysis at protein level. There was no statistically significant difference between women with RIF and fertile controls regarding PROK2 and PROKR2 levels. Altered expression of the PROK1/PROKR1 system could be one of the numerous abnormalities in the endometrium of women with RIF.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/metabolismo , Fertilização in vitro , Hormônios Gastrointestinais/genética , Expressão Gênica/fisiologia , Receptores Acoplados a Proteínas G/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Adulto , Endométrio/química , Feminino , Hormônios Gastrointestinais/análise , Hormônios Gastrointestinais/fisiologia , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/terapia , Gravidez , RNA Mensageiro/análise , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/fisiologia , Falha de Tratamento , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/análise , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia
8.
J Cell Mol Med ; 24(5): 3242-3245, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31991505

RESUMO

Prokineticin 1 (PROK1) is a key regulator of embryo implantation and placentation, and its dysregulation is associated with pregnancy complications, such as pre-eclampsia and foetal growth restriction. We have previously shown that insulin strongly enhances the expression of PROK1 in human decidualizing stromal cells. Here, we demonstrate that dihydrotestosterone (DHT), but not testosterone, potentiates insulin to up-regulate PROK1 in these cells. However, the androgens alone do not influence the expression of PROK1. Our findings suggest that insulin and androgens both are involved in the regulation of PROK1 that could have implications for normal and pathological pregnancies.


Assuntos
Di-Hidrotestosterona/farmacologia , Endométrio/metabolismo , Hormônios Gastrointestinais/genética , Insulina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Adolescente , Adulto , Androgênios/genética , Biópsia , Células Cultivadas , Decídua/metabolismo , Decídua/patologia , Implantação do Embrião/genética , Endométrio/efeitos dos fármacos , Endométrio/crescimento & desenvolvimento , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Placentação/genética , Pré-Eclâmpsia , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/genética , Complicações na Gravidez/patologia , Cultura Primária de Células , Transdução de Sinais/genética , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Ativação Transcricional , Adulto Jovem
9.
Fish Shellfish Immunol ; 98: 236-244, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31953197

RESUMO

Astakine is a crucial factor in the proliferation and differentiation of hematopoietic stem cells and is directly involved in hematopoiesis in crustaceans. To assess the role of Astakine in the innate immune system of Scylla paramamosain, the immune responses in healthy and Astakine-inhibited S. paramamosain were investigated in the present study. The RNA transcripts of Astakine were widely distributed in all examined tissues, with significantly higher levels of expression in hemocytes of both healthy and challenged S. paramamosain with Vibrio alginolyticus and WSSV. When Astakine was knocked down by RNA interference technology, immune-related genes, including Janus kinase, prophenoloxidase, hemocyanin, ß-actin, myosin II essential light chain-like protein, signal transducer and activator of transcription, Relish, and C-type-lectin, were significantly down-regulated in hemocytes. The levels of phenoloxidaseactivity (PO), total hemocyte counts (THC) and hemocyte proliferation decreased significantly in hemocytes of Astakine-dsRNA treated S. paramamosain. After being challenged with V. alginolyticus and WSSV, the THC decreased significantly and the levels of hemocyte apoptosis increased significantly in Astakine-dsRNA treated S. paramamosain in comparison with those in infected groups without Astakine-dsRNA treatment. After being challenged with WSSV, the WSSV copies were significantly lower in Astakine-dsRNA treated groups than those in the WSSV infection group, which suggested that knockdown of Astakine was not conductive to WSSV replication and this might be associated with the decreasing THC. The results of survival analysis showed that the survival rate of V. alginolyticus or WSSV infected S. paramamosain decreased significantly following Astakine knockdown. These results suggested that RNA interference of Astakine might weaken the resistance of S. paramamosain to V. alginolyticus or WSSV infection. The weaken resistivity after knockdown Astakine might be related to the changes of important immune-related gene expression, THC, PO activity, proliferation and apoptosis of hemocytes.


Assuntos
Proteínas de Artrópodes/metabolismo , Braquiúros/microbiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Vibrio alginolyticus/fisiologia , Vírus da Síndrome da Mancha Branca 1/fisiologia , Animais , Apoptose , Proteínas de Artrópodes/genética , Braquiúros/imunologia , Braquiúros/virologia , Proliferação de Células , Resistência à Doença/genética , Regulação da Expressão Gênica/imunologia , Hemócitos/metabolismo , Hemócitos/patologia , Imunidade Humoral , Taxa de Sobrevida , Distribuição Tecidual , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Replicação Viral
10.
Mol Med Rep ; 20(3): 2468-2475, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31322191

RESUMO

Prokineticin­1 (PROK1) serves important roles in the pathogenesis of polycystic ovary syndrome (PCOS); however, the association between microRNA (miR)­28­5p and PROK1 remains unclear. In the present study, the roles of miR­28­5p and PROK1, and their interaction in PCOS were investigated. Rat ovary granule cells were transfected with miR­28­5p mimics, and PROK1 expression levels were measured by reverse transcription­quantitative PCR and western blotting. A dual­luciferase reporter assay was performed to determine the association between miR­28­5p and PROK1. Additionally, pcDNA­PROK1 was co­transfected into rat ovary granule cells with miR­28­5p mimics. Cell proliferation, apoptosis, cell cycle and the expression of signaling proteins were investigated using Cell Counting Kit­8 assays, 5­ethynyl­2'­deoxyuridine staining, flow cytometry and western blotting, respectively. PROK1 expression was suppressed in rat ovary granule cells by miR­28­5p mimics, but upregulated following transfection with miR­28­5p inhibitors. The dual­luciferase reporter assay revealed that miR­28­5p binds to the 3'­untranslated region of PROK1. Proliferation activity was increased in PROK1­overexpressing cells; this effect was eliminated by co­transfection with miR­28­5p mimics. PROK1­overexpressing rat ovary granule cells exhibited significantly suppressed cell apoptosis and a decreased number of cells in G1; miR­28­5p mimics reversed these effects. Western blotting revealed that the PI3K/AKT/mTOR signaling pathway was activated by PROK1. The present results suggested that miR­28­5p attenuated the progression of PCOS by targeting PROK1, which may promote the pathogenesis of PCOS via the PI3K/AKT/mTOR pathway, indicating that the miR­28­5p/PROK1 axis may be a potential therapeutic target for patients with PCOS.


Assuntos
Hormônios Gastrointestinais/genética , MicroRNAs/genética , Síndrome do Ovário Policístico/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Animais , Linhagem Celular , Proliferação de Células , Progressão da Doença , Regulação para Baixo , Feminino , Síndrome do Ovário Policístico/patologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima
11.
Eur J Histochem ; 63(1)2019 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-30838843

RESUMO

Vascular endothelial growth factor (VEGF), its inhibitory splice variant, VEGF165b and Endocrine Gland derived VEGF (EG-VEGF) have a controversial role in pituitary gland. We aim to study VEGF, VEGF165b and EG-VEGF expression in pituitary adenomas. A significant correlation was found between growth hormone (GH) and VEGF secretion (P=0.024). For prolactinomas, VEGF and prolactin expression, had a P-value of 0.02 for Kendall coefficient and a P-value of 0.043 for the Spearman coefficient. VEGF-mRNA amplification was detected in both tumor cells and folliculostellate cells. VEGF165b was positive in 16.66% of pituitary adenomas. EG-VEGF was significantly correlated with prolactin (P=0.025) and luteinizing hormone (P=0.028). Our data strongly support VEGF, VEGF165b and EG-VEGF as important players of pituitary adenomas tumorigenesis. Particular hormonal milieu heterogeneity, special vascular network with an unusual reactivity to tumor growth correlated with variability of VEGF, VEGF165b and EG-VEGF secretion may stratify pituitary adenomas in several molecular groups with a direct impact on therapy and prognosis.


Assuntos
Adenoma/metabolismo , Hormônios Hipofisários/análise , Neoplasias Hipofisárias/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Adenoma/genética , Adenoma/patologia , Adenoma Acidófilo/genética , Adenoma Acidófilo/metabolismo , Adenoma Acidófilo/patologia , Adenoma Basófilo/genética , Adenoma Basófilo/metabolismo , Adenoma Basófilo/patologia , Adenoma Cromófobo/genética , Adenoma Cromófobo/metabolismo , Adenoma Cromófobo/patologia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
12.
Prog Mol Biol Transl Sci ; 161: 149-179, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30711026

RESUMO

Prokineticins are two conserved small proteins (~8kDa), prokineticin 1 (PROK1; also called EG-VEGF) and prokineticin 2 (PROK2; also called Bv8), with an N-terminal AVITGA sequence and 10 cysteines forming 5 disulfide bridges. PROK1 and PROK2 bind to two highly related G protein-coupled receptors (GPCRs), prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Prokineticins and their receptors are widely expressed. PROK1 is predominantly expressed in peripheral tissues, especially steroidogenic organs, whereas PROK2 is mainly expressed in the central nervous system and nonsteroidogenic cells of the testes. Prokineticins signaling has been implicated in several important physiological functions, including gastrointestinal smooth muscle contraction, circadian rhythm regulation, neurogenesis, angiogenesis, pain perception, mood regulation, and reproduction. Dysregulation of prokineticins signaling has been observed in a variety of diseases, such as cancer, ischemia, and neurodegeneration, in which prokineticins signaling seems to be a promising therapeutic target. Based on the phenotypes of knockout mice, PROKR2 and PROK2 have recently been identified as causative genes for idiopathic hypogonadotropic hypogonadism, a developmental disorder characterized by impaired development of gonadotropin-releasing hormone neurons and infertility. In vitro functional studies with these disease-associated PROKR2 mutations uncovered some novel features for this receptor, such as biased signaling, which may be used to understand GPCR signaling regulation in general.


Assuntos
Doença , Saúde , Receptores Acoplados a Proteínas G/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Mutação/genética , Nociceptividade , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/química , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
13.
Biomed Pharmacother ; 109: 762-769, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30551529

RESUMO

OBJECTIVE: Pancreatic carcinoma (PC), one of the most prevalent and malignant tumors, has a poor prognosis and a high mortality rate. EG-VEGF, a vascular endothelial growth factor from endocrine glands, also termed as PROK1, has a high positive expression rate in PC tissues and is involved in the pathogenesis of various tumors. However, the expression and potential role of EG-VEGF in PC has not been thoroughly explored. The aim of this study was to better clarify the expression and potential role of EG-VEGF in pancreatic carcinoma. METHODS: Immunohistochemical staining, western blotting, and RT-qPCR analysis were performed to detect the EG-VEGF level in PC tissues and cells. Subsequently, two short hairpin RNA (shRNA) lentiviral expression vector, shPROK1-1/shPROK1-2, were transfected into PANC-1 and BxPC-3 PC cell lines. MTT assay was used to determine cell proliferation. Meanwhile, flow cytometry assay was conducted to measure cell cycle and cell apoptosis. The protein levels of PI3K/AKT/mTOR pathway-related genes were also determined by western blotting. RESULTS: EG-VEGF was aberrantly expressed in PC samples, as compared with paracancerous samples. Knockdown of PROK1 notably decreased the protein level of EG-VEGF, indicating a successful downregulation model of EG-VEGF. EG-VEGF silencing remarkably attenuated cell proliferation, while also induced G0/G1 arrest and magnified the extent of cell apoptosis. Further, EG-VEGF knockdown significantly inhibited PI3K/AKT/mTOR signaling pathway by downregulating p-PI3K, p-AKT, and p-mTOR levels. CONCLUSION: This study identified the high-expression of EG-VEGF in pancreatic carcinoma tissues and cells, and demonstrated that EG-VEGF silencing inhibits the proliferation of PC cells and promotes apoptosis via regulating PI3K/AKT/mTOR pathway. Thus, EG-VEGF may become an essential target for the therapy of pancreatic cancer in the future.


Assuntos
Proliferação de Células/fisiologia , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Idoso , Apoptose/fisiologia , Linhagem Celular , Feminino , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Transdução de Sinais/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/antagonistas & inibidores , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Neoplasias Pancreáticas
14.
Epigenomics ; 10(9): 1243-1257, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30212243

RESUMO

AIM: Decidualization is essential for embryo implantation and placental development. We aimed to obtain transcriptome and epigenome profiles for primary endometrial stromal cells (ESCs) and in vitro decidualized cells. MATERIALS & METHODS: ESCs isolated from human endometrial tissues remained untreated (D0), or decidualized for 4 days (D4) and 8 days (D8) in the presence of 8-bromo-cAMP and progesterone. RESULTS: Among the epigenetic modifications examined (DNA methylation, H3K27ac, H3K9me3 and H3K27me3), the H3K27ac patterns changed most dramatically, with a moderate correlation with gene expression changes, upon decidualization. Subsets of up- and down-regulated genes upon decidualization were associated with reciprocal changes of H3K27ac and H3K27me3 modifications at their promoter region, and were enriched with genes essential for decidualization such as WNT4, ZBTB16, PROK1 and GREB1. CONCLUSION: Our dataset is useful to further elucidate the molecular mechanisms underlying decidualization.


Assuntos
Decídua/metabolismo , Implantação do Embrião/genética , Epigênese Genética , Histonas/metabolismo , Placentação/genética , Células Cultivadas , Metilação de DNA , Decídua/citologia , Feminino , Hormônios Gastrointestinais/genética , Humanos , Proteínas de Neoplasias/genética , Gravidez , Regiões Promotoras Genéticas , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Células Estromais/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Proteína Wnt4/genética
15.
Fish Shellfish Immunol ; 77: 419-428, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29609030

RESUMO

Astakine is a cytokine-like factor containing a prokineticin domain, which directly participates in hematopoiesis and blood cell differentiation. In the present study, a novel Astakine gene was identified from Chinese mitten crab Eriocheir sinensis (designated as EsAst). The full-length cDNA of EsAst was of 1163 bp, consisting of a 5' untranslated region (UTR) of 120 bp, a 3' UTR of 656 bp, and an open reading frame (ORF) of 387 bp encoding a polypeptide of 128 amino acids. There were a signal peptide and a prokineticin domain with nine conserved cysteine residues in the deduced amino acid sequence of EsAst. EsAst shared higher similarity with Astakines from Penaeus monodon and Pacifastacus leniusculus, and it was closely clustered with the Astakine from shrimp P. monodon in the phylogenetic tree. The EsAst mRNA transcript was higher expressed in hemocytes and hepatopancreas. The relative expression level of EsAst in hemocytes was continuously increased from 1.5 to 48 h after Vibro anguillarum challenge compared that in the untreated control group. After Pichia pastoris GS115 challenge, the relative expression level of EsAst in hemocytes was also up-regulated. After rEsAst injection, ROS levels in HPT cells were also increased at 12 and 24 h, and the total hemocyte counts were also significantly increased at 6, 9, 12, and 24 h post rEsAst injection. The interference of EsAst expression with dsRNA injection could delay the recovery of hemocytes production post A. hydrophila stimulation. When mitochondrial complexes I was knock down by dsRNA, ROS levels were decreased and THCs were also decreased. Recovery of hemocyte production inducing by A. hydrophila stimulation and rEsAst injection were delayed with dsEsbc1 injection. When ROS levels were increased after RNAi of Lon protease, THCs were also increased. The expression levels of five genes (EsJNK, EsSTAT, EsPI3K, EsAKT1, EsP70S6K) involved in SAPK-JNK and mTOR signaling pathways were up-regulated at 12 and 24 h in rEsAst group and EsLon dsRNA group compared with that in EGFP dsRNA group, and were similar to the trend of ROS levels. These results collectively suggested that EsAst should be a novel Astakine to promote the production of hemocytes in a ROS-dependent way in E. sinensis.


Assuntos
Braquiúros/genética , Braquiúros/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/imunologia , Aeromonas hydrophila/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/imunologia , Sequência de Bases , Perfilação da Expressão Gênica , Filogenia , Pichia/fisiologia , Distribuição Aleatória , Alinhamento de Sequência , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/química , Vibrio/fisiologia
16.
J Cell Mol Med ; 22(1): 163-172, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28782224

RESUMO

Prokineticin 1 (PROK1), a hypoxia-regulated angiogenic factor, has emerged as a crucial regulator of embryo implantation and placentation. Dysregulation of PROK1 has been linked to recurrent pregnancy loss, pre-eclampsia, foetal growth restriction and preterm birth. These pregnancy complications are common in women with obesity and polycystic ovary syndrome, i.e. conditions associated with insulin resistance and compensatory hyperinsulinaemia. We investigated the effect of insulin on PROK1 expression during in vitro decidualization. Endometrial stromal cells were isolated from six healthy, regularly menstruating women and decidualized in vitro. Insulin induced a significant dose-dependent up-regulation of PROK1 on both mRNA and protein level in decidualizing endometrial stromal cells. This up-regulation was mediated by hypoxia-inducible factor 1-alpha (HIF1α) via the phosphatidylinositol 3-kinase (PI3K) pathway. Furthermore, we demonstrated that PROK1 did not affect the viability, but significantly inhibited the migration of endometrial stromal cells and the migratory and invasive capacity of trophoblast cell lines. This in vitro study provides new insights into the regulation of PROK1 by insulin in human decidualizing endometrial stromal cells, the action of PROK1 on migration of endometrial stromal cells, as well as migration and invasion of trophoblasts. We speculate that hyperinsulinaemia may be involved in the mechanisms by which PROK1 is linked to placenta-related pregnancy complications.


Assuntos
Decídua/citologia , Decídua/metabolismo , Hormônios Gastrointestinais/genética , Insulina/farmacologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Adolescente , Adulto , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Coriocarcinoma/patologia , Feminino , Hormônios Gastrointestinais/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Trofoblastos/citologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Cicatrização/efeitos dos fármacos , Adulto Jovem
17.
Dev Comp Immunol ; 81: 141-151, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29154857

RESUMO

Astakine1 was isolated as a hematopoietic cytokine in the freshwater crayfish Pacifastacus leniusculus. In this study we detect and compare 79 sequences in GenBank, which we consider to be possible astakine orthologs, among which eleven are crustacean, sixteen are chelicerate and 52 are from insect species. Available arthropod genomes are searched for astakines, and in conclusion all astakine sequences in the current study have a similar exon containing CCXX(X), thus potentially indicating that they are homologous genes with the structure of this exon highly conserved. Two motifs, RYS and YP(N), are also conserved among the arthropod astakines. A phylogenetic analysis reveals that astakine1 and astakine2 from P. leniusculus and Procambarus clarkii are distantly related, and may have been derived from a gene duplication occurring early in crustacean evolution. Moreover, a structural comparison using the Mamba intestinal toxin (MIT1) from Dendroaspis polylepis as template indicates that the overall folds are similar in all crustacean astakines investigated.


Assuntos
Artrópodes/genética , Astacoidea/genética , Dendroaspis/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Animais , Evolução Biológica , Bases de Dados de Ácidos Nucleicos , Especiação Genética , Hematopoese/genética , Peptídeos e Proteínas de Sinalização Intercelular , Estrutura Molecular , Peptídeos/genética , Filogenia , Domínios Proteicos/genética , Especificidade da Espécie , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
18.
Cancer Biomark ; 21(2): 345-354, 2018 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-29226856

RESUMO

BACKGROUND: The highest risk factor for mortality among malignant tumors is metastasis. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor which biological activity is mediated via two G protein-coupled receptors, prokineticin receptor1 (PROKR1) and PROKR2. Recent studies suggested that EG-VEGF expression is deregulated in multiple cancers including colorectal cancer (CRC). METHODS: Using distinctive CRC and peritoneal carcinomatosis (PC) cohorts and a corresponding control cohort, we determined the circulating levels of EG-VEGF and its in situ expression, and that of its related receptors. RESULTS: Circulating EG-VEGF levels were significantly increased in patients with metastatic PC compared to CRC and control patients (p< 0.05). Furthermore, according to clinicopathologic examinations, local EG-VEGF expression correlated with higher tumor and nodal stages (p< 0.001) of CRC. EG-VEGF and PROKR2 were highly expressed in colorectal primary lesions compared to positive controls. PROKR1 expression was lower and did not change in tumor specimens. Also, EG-VEGF and its receptor PROKR2 were differentially expressed in the colorectal primary lesions and in the control groups. CONCLUSION: Altogether these findings suggest that EG-VEGF/receptors system might be an important actor in the CRC progression into PC and might be involved in the ability of tumor cells to invade other organs. Circulating EG-VEGF could be proposed as a prognostic marker in human CRC and its progression into PC.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Peritoneais/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Adolescente , Adulto , Idoso , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Fatores de Risco , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Adulto Jovem
19.
J Physiol Pharmacol ; 68(3): 477-484, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28820403

RESUMO

The aim of the study was to assess the predictive value of vascular endothelial growth factor (VEGF), its soluble receptor - sVEGF-R1/sFlt1 and endocrine gland-derived vascular endothelial growth factor (EG-VEGF) concentrations in serum and follicular fluid (FF) for ovarian hyperstimulation syndrome (OHSS) in women undergoing controlled ovarian hyperstimulation (COH) protocols. Patients have been divided into 3 groups: control group on natural cycle, patients stimulated with GnRH agonist and patients stimulated with GnRH antagonist. The FF and serum concentrations of VEGF, EG-VEGF, sVEGF R1 and the expression of VEGF and EG-VEGF mRNA in GC in small and large follicles collected from patients were investigated. When we compared all patients in a trial, OHSS occurrence was correlated with higher level of sVEGF R1 and a lower level of VEGF in a follicular fluid from large follicles in a day of oocyte retrieval. The VEGF/sVEGF-R1 ratio for patients in COH groups, above which the risk of developing OHSS is very low (OR 0.1 (95% CI 0.01 - 0.29, P = 0.0006) was found to be 0.281 pg/ml, with AUC - 0.738, P = 0.042, (95% CI 0.656 - 0.82). High levels of sVEGF-R1 and low level of VEGF in FF on the day of oocyte retrieval correlate with OHSS regardless of the stimulation protocol.


Assuntos
Líquido Folicular/metabolismo , Recuperação de Oócitos , Síndrome de Hiperestimulação Ovariana/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Adulto , Feminino , Fertilização in vitro , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Menotropinas/farmacologia , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
20.
Dev Comp Immunol ; 72: 37-43, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28163091

RESUMO

The golden apple snail Pomacea canaliculata is an invasive pest originating from South America. It has already been found in Asia, the southern United States and more recently in the EU. Aiming to target the immune system of the snail as a way to control its spreading, we have developed organ-specific transcriptomes and looked for molecules controlling replication and differentiation of snail hemocytes. The prokineticin domain-containing protein Astakine 1 is the only cytokine known thus far capable of regulating invertebrate hematopoiesis, and we analyzed the transcriptomes looking for molecules containing a prokineticin domain. We have identified a prokineticin-like protein (PlP), that we called Pc-plp and we analyzed by real-time PCR (qPCR) its expression. In control snails, highest levels of Pc-plp were detected in the digestive gland, the ampulla (i.e., a hemocyte reservoir) and the pericardial fluid (i.e., the hematopoietic district). We tested Pc-plp expression after triggering hematopoiesis via multiple hemolymph withdrawals, or during bacterial challenge through LPS injection. In both cases a reduction of Pc-plp mRNA was observed. The multiple hemolymph withdrawals caused a significant decrease of Pc-plp mRNA in pericardial fluid and circulating hemocytes, while the LPS injection promoted the Pc-plp mRNA drop in anterior kidney, mantle and gills, organs that may act as immune barrier in molluscs. Our data indicate an important role for prokineticin domain-containing proteins as immunomodulators also in gastropods and their dynamic expression may serve as a biosensor to gauge the effectiveness of immunological interventions aimed at curtailing the spreading of the gastropod pest P. canaliculata.


Assuntos
Citocinas/metabolismo , Gastrópodes/imunologia , Hemócitos/imunologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Animais , Citocinas/genética , Regulação para Baixo , Hematopoese , Hemolinfa , Imunidade Inata , Imunomodulação , Lipopolissacarídeos/imunologia , Transcriptoma , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA