Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Nutr Cancer ; 76(3): 279-295, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38226887

RESUMEN

This study aims to build a prognostic model based on lactic acid metabolism-related genes (LMRGs) to predict survival outcomes and tumor microenvironment status of Hepatocellular carcinoma (HCC) patients. The model was used to calculate riskscores of clinical samples. Survival analysis and Cox regression analysis were conducted to verify the independence and reliability of the riskscore to determine its clinical significance in prognosis evaluation of HCC. Additionally, we conducted a comprehensive analysis of tumor mutation burden (TMB), immune cell infiltration, and gene set molecular function in the high- and low-risk groups. We obtained 134 LMRGs mainly involved in cellular calcium homeostasis and calcium signaling pathways. The LMRGs in the risk assessment model included PFKFB4, SLC16A3, ADRA2B, SLC22A1, QRFPR, and PROK1. This study discovered much shorter overall survival and median survival time of patients with higher riskscores when compared to those with lower riskscores. It was indicated that for independent prediction of patients' prognosis, the riskscore had a significant clinical value. A remarkable difference was also found regarding TMB between the two groups. Finally, cell experiments demonstrated that the knockout of PFKFB4 and SLC16A3 genes suppressed lactate. Our research demonstrated that the riskscore, established based on LMRGs, is a promising biomarker.


Asunto(s)
Carcinoma Hepatocelular , Hormonas Gastrointestinales , Neoplasias Hepáticas , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Humanos , Ácido Láctico , Carcinoma Hepatocelular/genética , Microambiente Tumoral/genética , Reproducibilidad de los Resultados , Neoplasias Hepáticas/genética , Pronóstico , Receptores Acoplados a Proteínas G , Fosfofructoquinasa-2
2.
Int J Med Sci ; 21(1): 27-36, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164347

RESUMEN

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.


Asunto(s)
Hormonas Gastrointestinales , Preeclampsia , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Embarazo , Femenino , Humanos , Placenta/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Transducción de Señal/genética , Trofoblastos , Preeclampsia/genética , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo
3.
Int J Mol Sci ; 24(16)2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37629120

RESUMEN

Wharton's jelly (WJ) contains mesenchymal stem cells (MSCs) exhibiting broad immunomodulatory properties and differentiation capacity, which makes them a promising tool for cellular therapies. Although the osteogenic, chondrogenic and adipogenic differentiation is a gold standard for proper identification of MSCs, it is important to elucidate the exact molecular mechanisms governing these processes to develop safe and efficient cellular therapies. Umbilical cords were collected from healthy, full-term deliveries, for subsequent MSCs (WJ-MSCs) isolation. WJ-MSCs were cultivated in vitro for osteogenic, chondrogenic, adipogenic and neurogenic differentiation. The RNA samples were isolated and the transcript levels were evaluated using NovaSeq platform, which led to the identification of differentially expressed genes. Expression of H19 and SLPI was enhanced in adipocytes, chondrocytes and osteoblasts, and NPPB was decreased in all analyzed groups compared to the control. KISS1 was down-regulated in adipocytes, chondrocytes, and neural-like cells compared to the control. The most of identified genes were already implicated in differentiation of MSCs; however, some genes (PROK1, OCA2) have not yet been associated with initiating final cell fate. The current results indicate that both osteo- and adipo-induced WJ-MSCs share many similarities regarding the most overexpressed genes, while the neuro-induced WJ-MSCs are quite distinctive from the other three groups. Overall, this study provides an insight into the transcriptomic changes occurring during the differentiation of WJ-MSCs and enables the identification of novel markers involved in this process, which may serve as a reference for further research exploring the role of these genes in physiology of WJ-MSCs and in regenerative medicine.


Asunto(s)
Hormonas Gastrointestinales , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Gelatina de Wharton , Humanos , Condrocitos , Adipocitos , Diferenciación Celular/genética , Osteoblastos , Factores Inmunológicos
4.
Int J Mol Sci ; 23(19)2022 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-36233195

RESUMEN

Leber's hereditary optic neuropathy (LHON) is a maternally transmitted disease caused by mitochondria DNA (mtDNA) mutation. It is characterized by acute and subacute visual loss predominantly affecting young men. The mtDNA mutation is transmitted to all maternal lineages. However, only approximately 50% of men and 10% of women harboring a pathogenic mtDNA mutation develop optic neuropathy, reflecting both the incomplete penetrance and its unexplained male prevalence, where over 80% of patients are male. Nuclear modifier genes have been presumed to affect the penetrance of LHON. With conventional genetic methods, prior studies have failed to solve the underlying pathogenesis. Whole exome sequencing (WES) is a new molecular technique for sequencing the protein-coding region of all genes in a whole genome. We performed WES from five families with 17 members. These samples were divided into the proband group (probands with acute onset of LHON, n = 7) and control group (carriers including mother and relative carriers with mtDNSA 11778 mutation, without clinical manifestation of LHON, n = 10). Through whole exome analysis, we found that many mitochondria related (MT-related) nuclear genes have high percentage of variants in either the proband group or control group. The MT genes with a difference over 0.3 of mutation percentage between the proband and control groups include AK4, NSUN4, RDH13, COQ3, and FAHD1. In addition, the pathway analysis revealed that these genes were associated with cofactor metabolism pathways. Family-based analysis showed that several candidate MT genes including METAP1D (c.41G > T), ACACB (c.1029del), ME3 (c.972G > C), NIPSNAP3B (c.280G > C, c.476C > G), and NSUN4 (c.4A > G) were involved in the penetrance of LHON. A GWAS (genome wide association study) was performed, which found that ADGRG5 (Chr16:575620A:G), POLE4 (Chr2:7495872T:G), ERMAP (Chr1:4283044A:G), PIGR (Chr1:2069357C:T;2069358G:A), CDC42BPB (Chr14:102949A:G), PROK1 (Chr1:1104562A:G), BCAN (Chr 1:1566582C:T), and NES (Chr1:1566698A:G,1566705T:C, 1566707T:C) may be involved. The incomplete penetrance and male prevalence are still the major unexplained issues in LHON. Through whole exome analysis, we found several MT genes with a high percentage of variants were involved in a family-based analysis. Pathway analysis suggested a difference in the mutation burden of MT genes underlining the biosynthesis and metabolism pathways. In addition, the GWAS analysis also revealed several candidate nuclear modifier genes. The new technology of WES contributes to provide a highly efficient candidate gene screening function in molecular genetics.


Asunto(s)
Hormonas Gastrointestinales , Atrofia Óptica Hereditaria de Leber , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , ADN Mitocondrial/genética , Femenino , Genes Modificadores , Estudio de Asociación del Genoma Completo , Humanos , Hidrolasas/genética , Masculino , Metiltransferasas/genética , Mutación , Atrofia Óptica Hereditaria de Leber/genética , Linaje , Penetrancia
5.
Biol Reprod ; 104(1): 181-196, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32997136

RESUMEN

Acquisition of endometrial receptivity for embryo implantation is one of the crucial processes during pregnancy and is induced mainly by progesterone and enhanced by conceptus signals. Prokineticin 1 (PROK1) is characterized as a secretory protein with diverse functions in various tissues, including the reproductive tract. PROK1, with its receptor PROKR1, are up-regulated in the porcine endometrium during implantation and in women's receptive endometrium and decidua. However, the function of PROK1 in embryo-maternal communication has still not been fully elucidated. Hence, we hypothesize that PROK1 is involved in endometrial receptivity development and implantation in pigs. In this study, using the porcine in vivo model of intrauterine infusions of estradiol-17ß (E2) and prostaglandin E2 (PGE2), we revealed that these hormones elevated endometrial expression of PROK1 and PROKR1 mRNA, respectively. Moreover, E2, acting synergistically with PGE2, increased PROKR1 protein expression. We also evidenced that PROK1-PROKR1 signaling induced expression of following genes and/or proteins CCN2, CDH13, FGF2, NFATC2, ANGPT1, ANGPT2, CDH1, MUC4, SPP1, IFNG, IL6, LIF, LIFR, TNF, TGFB3, and FGF9, as well as phosphorylation of PTK2 and secretion of IL6 and IL11 by endometrial explants in vitro. Ingenuity pathway analysis revealed that functions associated with the PROK1-regulated genes/proteins include cell-to-cell contact, cell attachment, migration and viability, differentiation of epithelial tissue, leukocyte migration, inflammatory response, angiogenesis, and vasculogenesis. Summarizing, our study suggests that PROK1 acts pleiotropically as an embryonic signal mediator that regulates endometrial receptivity by increasing the expression of the genes and proteins involved in implantation and pregnancy establishment in pigs.


Asunto(s)
Implantación del Embrión/fisiología , Endometrio/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Dinoprostona/farmacología , Endometrio/efectos de los fármacos , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Fosforilación/efectos de los fármacos , Embarazo , Transducción de Señal/efectos de los fármacos , Porcinos
6.
Cell Biochem Funct ; 39(2): 308-316, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32876972

RESUMEN

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.


Asunto(s)
Exosomas/metabolismo , Hormonas Gastrointestinales/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Exosomas/trasplante , Femenino , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Trasplante Heterólogo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/antagonistas & inhibidores , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética
7.
World J Surg Oncol ; 19(1): 302, 2021 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-34657605

RESUMEN

BACKGROUND: Prokineticin 1 (PROK1) was reported as an angiogenic factor, which is associated with tumor progression, cell invasion, and metastasis in colorectal cancer. Although the association between PROK1 expression in primary cancer lesion and patient prognosis was reported, it is unclear whether plasma PROK1 concentration may be a predictive factor in colorectal cancer patients. This study investigated the association between PROK1 concentration in plasma and prognosis in colorectal cancer patients. METHODS: We measured preoperative PROK1 plasma levels using ELISA method, while PROK1 expression in primary cancer lesion was evaluated using immunohistochemistry (IHC). The association between plasma PROK1 levels and cancer-related survival rate (CRS) was evaluated. Additionally, we examined whether simultaneous PROK1 expression in both primary cancer lesions and plasma was correlated with CRS. The cancer-related survival rate was calculated using the Kaplan-Meier method, and survival estimates were compared using the log-rank test. RESULTS: We have gathered eligible 130 CRC patients retrospectively. Out of 130 patients, 61 (46.9%) were positive on IHC in primary cancer, and 69 (53.1%) were negative, while 43 (33.1%) had high-value PROK1 in plasma. Out of these 43, 30 (25.4%) also had concomitant higher IHC expression in primary cancer. The plasma PROK1 levels tended to increase with advancing stages. The plasma PROK1-positive group had a lower 5-year CRS than the negative group (63.6% vs. 88.2%; P = 0.006). Additionally, simultaneous PROK1 expression was associated with a more significant decrease of 5-year CRS than both negative groups in all stages (76.2% vs. 92.5%; P = 0.003) and stage III (59.3% vs. 84.5%; P = 0.047). Multivariate analysis showed simultaneous PROK1 expression was independently associated with worse CRS (HR, 1.97; 95% CI 1.20­3.24, P < 0.01). CONCLUSION: PROK1 expression in preoperative plasma reflects poor prognosis in patients undergoing curative resection for colorectal cancer. The plasma PROK1 level may be a potential predictive marker, especially in stage III colorectal cancer patients.


Asunto(s)
Neoplasias Colorrectales , Hormonas Gastrointestinales , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Biomarcadores/sangre , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/cirugía , Hormonas Gastrointestinales/sangre , Humanos , Pronóstico , Estudios Retrospectivos , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/sangre
8.
Acta Med Okayama ; 75(6): 677-684, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34955534

RESUMEN

Extravillous trophoblast (EVT) invasion is important for embryo implantation, placental development, and successful remodeling of the uterine spiral artery. Endocrine gland derived-vascular endothelial growth factor (EG-VEGF) and matrix metalloproteinases (MMPs) are implicated in EVT invasion; however, the high con-centrations found in pregnancy pathologies have not been investigated in non-tumor trophoblasts. The roles of EG-VEGF, prokineticin receptors (PROKR1/2), MMP-2, and MMP-9 in EVT invasion during spiral artery remodeling were evaluated using human EVT from HTR-8/SVneo cell lines. The expression of MMP-2, MMP-9, and mitogen-activated protein kinase (MAPK), and Akt pathways in HTR-8/SVneo cells treated with recom-binant EG-VEGF alongside anti-PROKR1 and/or anti-PROKR2 antibodies was evaluated using quantitative reverse transcription-PCR and western blotting. Wound-healing and cell invasion assays were performed to assess the migration and invasion of these treated cells. Interestingly, 20 nM EG-VEGF activated ERK1/2 sig-naling and upregulated MMP-2 and MMP-9. This effect was suppressed by anti-PROKR2 antibody via ERK1/2 downregulation. Anti-PROKR2 antibody inhibited the migration and invasion of EG-VEGF-stimulated HTR-8/SVneo cells. Elevated concentrations of EG-VEGF enhance EVT invasion in a human trophoblast cell line by upregulating MMP-2 and MMP-9 via PROKR2. These new insights into the regulation of epithelial cell invasion may help in developing therapeutic interventions for placental-related diseases during pregnancy.


Asunto(s)
Trofoblastos/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Línea Celular , Femenino , Humanos , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Placenta/metabolismo , Embarazo , Receptores Acoplados a Proteínas G , Receptores de Péptidos/metabolismo , Transducción de Señal , Regulación hacia Arriba , Arteria Uterina
9.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638592

RESUMEN

The development of neuropathy and of mood alterations is frequent after chemotherapy. These complications, independent from the antitumoral mechanism, are interconnected due to an overlapping in their processing pathways and a common neuroinflammatory condition. This study aims to verify whether in mice the treatment with the proteasome inhibitor bortezomib (BTZ), at a protocol capable of inducing painful neuropathy, is associated with anxiety, depression and supraspinal neuroinflammation. We also verify if the therapeutic treatment with the antagonist of the prokineticin (PK) system PC1, which is known to contrast pain and neuroinflammation, can prevent mood alterations. Mice were treated with BTZ (0.4 mg/kg three times/week for 4 weeks); mechanical allodynia and locomotor activity were evaluated over time while anxiety (dark light and marble burying test), depression (sucrose preference and swimming test) and supraspinal neuroinflammation were checked at the end of the protocol. BTZ treated neuropathic mice develop anxiety and depression. The presence of mood alterations is related to the presence of neuroinflammation and PK system activation in prefrontal cortex, hippocampus and hypothalamus with high levels of PK2 and PKR2 receptor, IL-6 and TNF-α, TLR4 and an upregulation of glial markers. PC1 treatment, counteracting pain, prevented the development of supraspinal inflammation and depression-like behavior in BTZ mice.


Asunto(s)
Afecto/efectos de los fármacos , Bortezomib/farmacología , Inhibidores de Proteasoma/farmacología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Biomarcadores/metabolismo , Citocinas/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/tratamiento farmacológico , Dolor/metabolismo , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
J Cell Mol Med ; 24(5): 3242-3245, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31991505

RESUMEN

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.


Asunto(s)
Dihidrotestosterona/farmacología , Endometrio/metabolismo , Hormonas Gastrointestinales/genética , Insulina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Adolescente , Adulto , Andrógenos/genética , Biopsia , Células Cultivadas , Decidua/metabolismo , Decidua/patología , Implantación del Embrión/genética , Endometrio/efectos de los fármacos , Endometrio/crecimiento & desarrollo , Células Epiteliales/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Placentación/genética , Preeclampsia , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/patología , Cultivo Primario de Células , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Activación Transcripcional , Adulto Joven
11.
Biol Reprod ; 103(3): 654-668, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32355954

RESUMEN

Pregnancy establishment in mammals, including pigs, requires proper communication between embryos and the maternal reproductive tract. Prokineticin 1 (PROK1) has been described as a secretory protein with pleiotropic functions and as a novel tissue-specific angiogenic factor. However, despite the studies performed mainly on human cell lines and in mice, the function of PROK1 in the endometrium during early pregnancy is still not fully elucidated. We hypothesized that PROK1 contributes to pregnancy establishment in pigs. The present study is the first to report that the expression of PROK1 and its receptor (PROKR1) is elevated in the porcine endometrium during the implantation and early placentation period. PROK1 protein was detected mainly in luminal epithelial cells, glandular epithelial cells, and blood vessels in the endometrium. Using the porcine in vivo model of unilateral pregnancy, we revealed that conceptuses induced the endometrial expression of PROK1 and PROKR1. Moreover, the embryonic signal, estradiol-17ß, as well as progesterone, stimulated the endometrial expression of PROK1 and PROKR1. We also evidenced that PROK1-PROKR1 signaling supports endometrial angiogenesis in pigs. The PROK1-stimulated proliferation of primary porcine endometrial endothelial (PEE) cells involved PI3K/AKT/mTOR, MAPK, cAMP, and NFKB signaling pathways. Furthermore, PROK1 via PROKR1 promoted the formation of capillary-like structures by PEE cells. PROK1 also stimulated VEGFA and PGF2α secretion, which in turn may indirectly support angiogenic changes within endometrial tissue. In summary, our study suggests that PROK1 acts as an embryonic signal mediator that regulates endometrial angiogenesis and secretory function during the implantation and early placentation period in pigs.


Asunto(s)
Endometrio/metabolismo , Neovascularización Fisiológica/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/fisiología , Animales , Proliferación Celular , Células Endoteliales/metabolismo , Femenino , Feto/metabolismo , Inmunohistoquímica , Embarazo , Porcinos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
12.
Pharmacol Res ; 160: 105190, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32937177

RESUMEN

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.


Asunto(s)
Encefalopatías/genética , Encéfalo/crecimiento & desarrollo , Cardiopatías/genética , Corazón/crecimiento & desarrollo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/fisiología , Animales , Encéfalo/fisiología , Corazón/fisiología , Humanos , Neurogénesis/genética , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología
13.
Fish Shellfish Immunol ; 98: 236-244, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31953197

RESUMEN

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.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Braquiuros/microbiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Vibrio alginolyticus/fisiología , Virus del Síndrome de la Mancha Blanca 1/fisiología , Animales , Apoptosis , Proteínas de Artrópodos/genética , Braquiuros/inmunología , Braquiuros/virología , Proliferación Celular , Resistencia a la Enfermedad/genética , Regulación de la Expresión Génica/inmunología , Hemocitos/metabolismo , Hemocitos/patología , Inmunidad Humoral , Tasa de Supervivencia , Distribución Tisular , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Replicación Viral
14.
Clin Otolaryngol ; 45(5): 788-795, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32516473

RESUMEN

OBJECTIVE: To characterise the role of VEGF, EG-VEGF and its receptors in the development and progression of HNC. DESIGN: Human serum and tissues samples were collected from healthy, epulis and HNC patients and used for ELISA assays and immunohistochemistry studies, respectively. SETTING: Ibn Rochd Hospital of Casablanca (Morocco), INSERM and University of Grenoble Alpes (France). PARTICIPANTS: We used serum from 64 patients with head and neck cancers and from 71 controls without general pathology. Tissues samples were collected from seven patients with OSCC and from seven patients with Epulis. MAIN OUTCOME MEASURES: We compared circulating VEGF and EG-VEGF in normal and HNC patients and determined the expression, localisation and quantification of VEGF, EG-VEGF and its receptors; PROKR1 and PROKR2 as well as Ki67, CD31 and CD34 in OSCC and Epulis patients. RESULTS: Both EG-VEGF and VEGF circulating levels were significantly decreased in the HNC (P < .01). OSCC patients expressed less EG-VEGF and VEGF proteins, higher PROKR1 and PROKR2 with no change in CD31 and CD34 levels. A significant increase in Ki67 was observed in OSCC. CONCLUSIONS: We demonstrated that circulating VEGF and EG-VEGF are downregulated in HNC patients and in OSCC tissue. EG-VEGF receptors were increased in OSCC, along with a stabilisation of two key markers of angiogenesis. These findings strongly suggest that downregulation of angiogenesis in HNC might explain its moderate metastatic feature.


Asunto(s)
Regulación hacia Abajo , Neoplasias de Cabeza y Cuello/sangre , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/biosíntesis , Adulto , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/sangre , Progresión de la Enfermedad , Glándulas Endocrinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Masculino , Factor A de Crecimiento Endotelial Vascular/sangre , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/sangre , Adulto Joven
15.
J Physiol ; 597(12): 3069-3083, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31026335

RESUMEN

KEY POINTS: Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is a critical factor that facilitates trophoblast invasion in placenta. Plasma miR-141 and miR-200a levels were elevated, while EG-VEGF was decreased in peripheral blood and placenta of preeclamptic patients. Furthermore, numbers of cilia in the placenta from preeclamptic women were significantly decreased. Elevated miR-141 and miR-200a inhibited the expression of EG-VEGF, downstream extracellular signal-regulated kinase (ERK)/matrix metalloproteinase 9 signalling and cilia formation, thus leading to defective trophoblast invasion. The growth of the primary cilium, which transduced ERK signalling upon EG-VEGF induction for proper trophoblast invasion, was also inhibited by miR-141 and miR-200a upregulation. ABSTRACT: Preeclampsia is a severe gestational complication, and inadequate trophoblast invasion during placental development is an important pathoaetiology. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is a critical factor that facilitates trophoblast invasion in placenta. By binding to the primary cilium, EG-VEGF initiates the signalling cascade for proper embryo implantation and placental development. The miR-200 family was predicted to target the EG-VEGF 5'-untranslated region, and its specific binding site was confirmed using a dual luciferase and a co-transfection assay. In the peripheral blood and placenta of preeclamptic patients, EG-VEGF showed significantly lower expression, whereas plasma miR-141 and miR-200a had higher expression compared with the controls. The biological significance of miR-141 and miR-200a was verified using an overexpression method in a trophoblast cell line (HTR-8/SVneo). Elevated miR-141 and miR-200a inhibited the expression of EG-VEGF, matrix metalloproteinase 9 (MMP9) and downstream extracellular signal-regulated kinase (ERK) signalling, thus leading to defective trophoblast invasion. Additionally, the growth of the primary cilium, which transduces ERK/MMP9 signalling upon EG-VEGF induction, was inhibited by miR-141 and miR-200a upregulation. Furthermore, the number of cilia in the human placenta of preeclamptic women was significantly decreased compared to normal placenta. In conclusion, the study uncovers the clinical correlations among the miR-200 family, EG-VEGF and the primary cilium in preeclampsia and the underlying molecular mechanisms. The results indicate that miR-141 and miR-200a directly targeted EG-VEGF, suppressed primary cilia formation and inhibited trophoblast invasion. Thus, miR-141 and miR-200a could be explored as promising miRNA biomarkers and therapeutic targets in preeclampsia.


Asunto(s)
Cilios/fisiología , MicroARNs/sangre , Preeclampsia/sangre , Adulto , Biomarcadores/sangre , Línea Celular , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Recién Nacido , Masculino , Preeclampsia/genética , Preeclampsia/fisiopatología , Embarazo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/sangre
16.
Biol Reprod ; 101(4): 832-841, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31276578

RESUMEN

CONTEXT: Prokineticin 1 (PROK1) quantification in global follicular fluid (FF) has been recently reported as a predictive biomarker of in vitro fertilization (IVF) outcome. It is now necessary to evaluate its clinical usefulness in individual follicles. OBJECTIVES: To evaluate the clinical value of PROK1 secretion in individual FF to predict oocyte competence. To determine the impact of follicular size, oocyte maturity, and gonadotropin treatments on PROK1 secretion. DESIGN AND SETTING: Prospective cohort study from May 2015 to May 2017 at the University Hospital of Grenoble. PATIENTS: A total of 69 infertile couples underwent IVF. INTERVENTION(S): Collection of 298 individual FF from 44 women undergoing IVF; 52 individual cumulus cell (CC) samples and 15 CC primary cultures from 25 women undergoing IVF-intracytoplasmic sperm injection (ICSI). MAIN OUTCOME MEASURE(S): Oocyte competence was defined as the ability to sustain embryo development to the blastocyst stage. Follicular size was measured by 2D-sonography. PROK1 concentration was quantified by ELISA assay. RESULTS: PROK1 concentration was correlated to follicular size (r = 0.85, P = 2.2 × 10-16). Normalized PROK1 concentration in FF was predictive of subsequent oocyte competence (AUROC curve = 0.76 [95% CI, 0.69-0.83]; P = 1.7 × 10-9), irrespectively of day-2 embryo morphokinetic parameters. The expression and secretion of PROK1 were increased in FF and CC of mature oocytes (P < 0.01). Follicle Stimulating Hormone and hCG up-regulated PROK1 secretion in CC primary cultures (P < 0.01; P < 0.05), probably through the cAMP pathway (P < 0.01). CONCLUSIONS: PROK1 quantification in individual FF could constitute a new predictive biomarker of oocyte competence in addition with embryo morphokinetic parameters. TRIAL REGISTRATION NUMBER: none.


Asunto(s)
Biomarcadores/análisis , Desarrollo Embrionario , Líquido Folicular/química , Hormonas Gastrointestinales/análisis , Oocitos/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/análisis , Biomarcadores/metabolismo , Células Cultivadas , Estudios de Cohortes , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Fertilización In Vitro , Líquido Folicular/metabolismo , Francia , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo , Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Humanos , Recuperación del Oocito/normas , Oocitos/citología , Oogénesis/efectos de los fármacos , Oogénesis/genética , Oogénesis/fisiología , Embarazo , Índice de Embarazo , Pronóstico , Estudios Prospectivos , Control de Calidad , Inyecciones de Esperma Intracitoplasmáticas , Resultado del Tratamiento , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
17.
Fish Shellfish Immunol ; 94: 66-71, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31465872

RESUMEN

Astakine 1 is a small cytokine-like peptide which is directly involved in hematopoiesis in crustaceans. Astakines are present in many different invertebrate groups primarily in arthropods. In this study we found that astakine1 was present as a high molecular weight (HMW) complex in plasma. It is known that calcium concentration are fluctuating in several crustaceans especially during the molting process. This HMW-complex was formed under low calcium concentrations in plasma and could be partially reversed provided calcium was added. The biological function of the naïve astakine1 and that in the HMW complex was about the same, but if the protein is to be isolated or studied for its function it is important to know about this property of astakine1 which may previously have hampered isolation and functional studies in other animals than freshwater crayfish.


Asunto(s)
Proteínas de Artrópodos/genética , Astacoidea/genética , Astacoidea/inmunología , Calcio/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/inmunología , Animales , Proteínas de Artrópodos/inmunología , Plasma/química
18.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2322-2329, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28939058

RESUMEN

The Melanocortin Receptor Accessory Protein 2 (MRAP2) regulates the activity of several GPCRs involved in the control of food intake and energy expenditure. While MRAP2 was originally thought to exclusively interact with melanocortin receptors we have recently shown that it interacts with and inhibits the trafficking and signaling of the prokineticin receptor 1 (PKR1). In this study we demonstrate a new role of MRAP2 in the regulation of the orexin receptor 1 (OX1R) and identify the specific regions of MRAP2 required for the regulation of OX1R and PKR1. Importantly, like MC4R and PKRs, OX1R is predominately expressed in the brain where it regulates food intake. By demonstrating that MRAP2 modulates the activity of OX1R we further establish the critical role of MRAP2 in the control of energy homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Ingestión de Alimentos/genética , Receptores de Orexina/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Señalización del Calcio/genética , Proteínas Portadoras/genética , Cricetulus , Metabolismo Energético/genética , Humanos , Orexinas/genética , Orexinas/metabolismo , Transducción de Señal , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
19.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 44(6): 621-627, 2019 Jun 28.
Artículo en Zh | MEDLINE | ID: mdl-31304922

RESUMEN

OBJECTIVE: To investigate the role of prokineticin (PROK) 1 and prokineticin-receptor (PROKR) 1 in the pathogenesis of endometriosis and its clinical signifaicance.
 Methods: Quantitative real-time PCR (qPCR) and Western bloting were used to detect the expression of PROK 1 and PROKR 1 in eutopic and ectopic endometrium of endometriosis (n=22) and normal control endometrium (n=18). Endometrial stromal cells were isolated and cultured in 6 normal controls. The expression of PROK 1 mRNA was detected by qPCR after treated with estradiol (E2) or TNF-α.
 Results: PROK 1 and PROKR 1 mRNA were expressed in eutopic and ectopic endometrium of endometriosis and normal control endometrium, and the expression level gradually declined (P<0.05). The expression of PROKR-1 protein in eutopic and ectopic endometrium of endometriosis and normal control endometrium gradually declined (P<0.05). The expression of PROK-1 protein in normal control endometrial cells and eutopic endometrium cell was higher in secretory phase than in proliferative phase (P<0. 05). E2 did not change the expression of PROK 1, whereas TNF-α up-regulated the expression of PROK 1.
 Conclusion: PROK-1 and its receptors are involved in the pathogenesis and development of endometriosis. TNF-α can promote angiogenesis via up-regulating the expression of PROK 1.


Asunto(s)
Endometriosis , Hormonas Gastrointestinales/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Endometrio , Femenino , Humanos , ARN Mensajero
20.
J Cell Mol Med ; 22(1): 163-172, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782224

RESUMEN

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.


Asunto(s)
Decidua/citología , Decidua/metabolismo , Hormonas Gastrointestinales/genética , Insulina/farmacología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Adolescente , Adulto , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/patología , Femenino , Hormonas Gastrointestinales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Trofoblastos/citología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA