Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Clin Otolaryngol ; 45(5): 788-795, 2020 09.
Article in English | MEDLINE | ID: mdl-32516473

ABSTRACT

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.


Subject(s)
Down-Regulation , Head and Neck Neoplasms/blood , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Adult , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/blood , Disease Progression , Endocrine Glands/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/blood , Young Adult
2.
Biomed Pharmacother ; 109: 762-769, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551529

ABSTRACT

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.


Subject(s)
Cell Proliferation/physiology , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Aged , Apoptosis/physiology , Cell Line , Female , Gene Silencing , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/antagonists & inhibitors , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Pancreatic Neoplasms
3.
Int J Clin Exp Pathol ; 8(8): 8902-11, 2015.
Article in English | MEDLINE | ID: mdl-26464631

ABSTRACT

OBJECTIVE: To investigate the effect of ovarian stimulation on the expression of EG-VEGF mRNA and protein in peri-implantation endometrium in women undergoing IVF and its relation with endometrial receptivity (ER). DESIGN: Prospective laboratory study. SETTING: University hospital. PATIENTS: Eighteen women in stimulated cycles (SC) as study subjects and 18 women in natural cycles (NC) as controls. Women in SC group were classified with two subgroups, high ovarian response (SC1, n=9) with peak serum E2>5,000 pg/mL and moderate ovarian response (SC2, n=9) with peak serum E2 1,000-5,000 pg/mL. INTERVENTION(S): Endometrial biopsies were collected 6 days after ovulation in NC or after oocyte retrieval in SC. MAIN OUTCOME MEASURE(S): Endometrium histological dating was observed with HE staining. EG-VEGF mRNA expression levels determined by real-time polymerase chain reaction analysis, and protein levels by immunohistochemistry. RESULTS: All endometrial samples were in the secretory phase. The endometrial development in SC1 was 1 to 2 days advanced to NC, and with dyssynchrony between glandular and stromal tissue. Immunohistochemistry analysis showed that EG-VEGF protein was predominantly expressed in the glandular epithelial cells and endothelial cells of vessels, and also presented in the stroma. The image analysis confirmed that both the gland and stroma of endometrium in SC1 had a significantly lower EG-VEGF protein expression than that in SC2 and NC endometrium. Moreover, EG-VEGF mRNA levels were significantly lower in SC1 than in NC. Both EG-VEGF protein and mRNA levels had no significant difference between SC2 and NC. CONCLUSION: Decreased expression of EG-VEGF in the peri-implantation is associated with high ovarian response, which may account for the impaired ER and lower implantation rate in IVF cycles.


Subject(s)
Endometrium/metabolism , Fertilization in Vitro , Ovulation Induction , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Adult , Female , Fertilization in Vitro/methods , Humans , Immunohistochemistry , Ovulation Induction/methods , Prospective Studies , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction
4.
Rev Med Chir Soc Med Nat Iasi ; 119(2): 461-5, 2015.
Article in English | MEDLINE | ID: mdl-26204653

ABSTRACT

AIM: To prove the presence of EG-VEGF in tumor ovary and to analyze its involvement in the ovarian carcinogenesis, as promoter of angiogenesis, in relationship with the clinicopathological prognostic factors and survival. METHODS: The study group comprises tumor tissue specimens from 50 cases of surgically treated ovarian cancer that were immunohistochemically investigated. A scoring system based on the percentage of positive cells and the intensity of staining was applied for the semiquantitative assessment of EG-VEGF, as negative or positive. Statistics involved χ2 test, and Kaplan-Meier and log-rank test. RESULTS: EG-VEGF was positive in 35 cases (70%) and negative in 15 cases (30%). Our data confirmed the predominance of EG-VEGF positivity in the serous subiype as compared to endometrioid and clear cell subtypes, and its absence in mucinous subtype. Moreover, we demonstrated that EG-VEGF is overexpressed mainly in high-grade ovarian carcinomas (type II) than in low-grade ones. Significant differences were registered between the EG-VEGF positive or negative expression and tumor stage and histological subtypes, respectively. Survival analysis showed no differences in patient's survival and EG-VEGF positive and negative cases. CONCLUSIONS: The analysis of EG-VEGF expression in ovarian tumors points out the relationship between the enhanced potential for tumor angiogenesis and the tumor aggressivity.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Adult , Aged , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/surgery , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Predictive Value of Tests , Prognosis , Sensitivity and Specificity
5.
Oncotarget ; 6(8): 6053-61, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25788276

ABSTRACT

Hematogenous metastasis, mainly hepatic metastasis, is a frequent metastatic mode in colorectal cancer involving angiogenic growth factors. Two angiogenic growth factors, in particular, Vascular endothelial growth factor (VEGF) and Prokineticin1(PROK1), are considered to have an important role in hematogenous metastasis of colorectal cancer. Accordingly, we report our findings on the importance of the anti-tumor efffect by inhibiting these two factors in human colorectal cancer.When the culture fluid of Colorectal cancer cell lines(DLD-1, HCT116, and LoVo) with high levels of VEGF/PROK1 expression was injected subcutaneously into mice, the culture fluid increased subcutaneous angiogenesis. But when both anti-PROK1 and anti-VEGF antibodies were present in the culture fluid, the length and size of the blood vessels were reduced compared with those seen in the fluid-only, anti-PROK1, and anti-VEGF controls. Also, tumor masses were produced in mice by subcutaneously embedding colorectal cancer cells with high levels VEGF/PROK1 expression. When both anti-PROK1 and anti-VEGF antibodies were simultaneously applied, tumor formation and peritumoral angiogenesis were strongly suppressed, compared with when either anti-PROK1 antibody or anti-VEGF antibody was applied alone.Simultaneous targeting of both angiogenic growth factors (VEGF/PROK1) may prove more useful in colorectal cancer.


Subject(s)
Antibodies/pharmacology , Colorectal Neoplasms/therapy , Gastrointestinal Hormones/immunology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/immunology , Animals , Cell Line, Tumor , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Female , Gastrointestinal Hormones/biosynthesis , HCT116 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Xenograft Model Antitumor Assays
6.
PLoS One ; 8(8): e72793, 2013.
Article in English | MEDLINE | ID: mdl-24013515

ABSTRACT

Astakine is an important cytokine that is involved in crustacean hematopoiesis. Interestingly, the protein levels of astakine increased dramatically in plasma of LPS-injected shrimp while mRNA levels remained unchanged. Here, we investigated the involvement of astakine 3'-untranslated region (UTR) in its protein expression. The 3'-UTR of astakine down-regulated the expression of reporter protein but the mRNA stability of reporter gene was unaffected. We identified the functional regulatory elements of astakine 3'-UTR, where 3'-UTR242-483 acted as repressor. The electrophoresis mobility shift assay (EMSA), RNA pull-down assay and LC/MS/MS were performed to identify the protein association. We noted that crustin Pm4 and shrimp transglutaminase I (STG I) were associated to astakine 3'-UTR242-483, while two other proteins have yet to be revealed. Depletion of hemocytic crustin Pm4 and STG I significantly increased the protein level of astakine while astakine mRNA level remained unaffected. Lipopolysaccharide (LPS) stimulated the secretion of crustin Pm4 and STG I from hemocytes to plasma and increased the astakine level to stimulate the hemocytes proliferation. Altogether, we identified the shrimp crustin Pm4 and STG I as novel RNA binding proteins that play an important role in down-regulating astakine expression at post-transcriptional level and are crucial for the maintenance of hematopoiesis.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Arthropod Proteins/metabolism , Decapoda/metabolism , Down-Regulation/physiology , Hematopoiesis/physiology , Ribonucleoproteins/metabolism , Transglutaminases/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , 3' Untranslated Regions/physiology , Animals , Antimicrobial Cationic Peptides/genetics , Arthropod Proteins/genetics , Decapoda/genetics , Hemocytes/cytology , Hemocytes/metabolism , RNA Stability/physiology , Ribonucleoproteins/genetics , Sf9 Cells , Spodoptera , Transglutaminases/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
7.
Rom J Morphol Embryol ; 53(3): 479-83, 2012.
Article in English | MEDLINE | ID: mdl-22990536

ABSTRACT

Ovarian cancer-related angiogenesis is a complex process orchestrated by many positive and negative regulators. Many growth factors are involved in the development of the tumor-associated vasculature, and from these, endocrine gland-derived vascular endothelial growth factor (EG-VEGF) seems to play a crucial role. EG-VEGF is the first organ-specific angiogenic factor and its effects are restricted to the endothelial cells of the endocrine glands. Although EG-VEGF was detected in both normal and neoplastic ovaries, its clinical significance remains controversial. In the present study, we analyzed 30 patients with epithelial ovarian cancer, and the immunohistochemical expression of EG-VEGF was compared with the conventional clinico-pathological parameters of prognosis. Neoplastic cells of the ovarian carcinoma expressed EG-VEGF in 73.33% of the cases, as a cytoplasmic granular product of reaction. We found a strong correlation between the expression of EG-VEGF at protein level and tumor stage, grade, and microscopic type. The expression of EG-VEGF was found in patients with stage III and IV, but not in stage II. The majority of serous adenocarcinoma, half of the cases with clear cell carcinoma and two cases with endometrioid carcinoma showed definite expression in tumor cells. No positive reaction was found in the cases with mucinous carcinoma. Our results showed that EG-VEGF expression is an indicator not only of the advanced stage, but also of ovarian cancer progression. Based on these data, we concluded that EG-VEGF expression in tumor cells of the epithelial ovarian cancer is a good marker of unfavorable prognosis and could be an attractive therapeutic target in patients with advanced-stage tumors, refractory conventional chemotherapy.


Subject(s)
Biomarkers, Tumor/biosynthesis , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Aged , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/pathology , Prognosis , Treatment Outcome , Tumor Cells, Cultured
8.
Thyroid ; 21(4): 391-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21385081

ABSTRACT

BACKGROUND: Endocrine gland-derived vascular endothelial growth factor (Prok1) and prokineticin 2 (Prok2) are involved in the organ-specific regulation of angiogenesis, which is a crucial step toward cancer progression in most tumors, including those of thyroid gland. The oncogene BRAF V600E mutation is associated with poor clinical outcome of papillary thyroid cancer (PTC) and can independently predict its recurrence. DESIGN: Our hypothesis was that Prok1 and Prok2 expression levels associated with BRAF mutations can be prognostic factors for PTC outcome. Prok1 and Prok2 were examined in PTC, a cell line derived from a human PTC (designated FB-2), euthyroid multinodular goiter (MNG), Graves' disease (GD), and contralateral normal thyroid (NT) tissues from PTC cases. We evaluated BRAF mutation and its relationship with Prok1 expression pattern in PTC. METHODS: We studied Prok1 and Prok2 mRNAs by real-time polymerase chain reaction and BRAF mutation by mutant allele-specific polymerase chain reaction amplification. Formalin-fixed, paraffin-embedded blocks of PTC and NT were used for the immunohistochemical determination of Prok1 using anti-endocrine gland vascular endothelial growth factor primary antibody. RESULTS: Prok1 and Prok2 transcripts were both present in thyroid tissues, and Prok1 was differentially expressed in PTC compared to MNG, GD, and NT. Prok1 mRNA levels were very low in NT and MNG and significantly higher in PTC, FB-2, and GD (p<0.05). Prok1 protein was almost undetectable in NT but was highly expressed in all PTC samples having an infiltrative pattern of growth and lymph node metastases ( p<0.05). Further, the expression of Prok1 in PTC was associated with 60% of the samples being positive for the BRAF mutation ( p<0.05). CONCLUSIONS: We found that Prok1 is significantly increased in PTC, and its expression in PTC is related to BRAF mutation. These results suggest that Prok1 could be a new useful marker for thyroid cancer progression. Prok1 therefore could also be a potential target for novel therapeutic strategies, although the lack of functional data suggests caution against generalization of this assumption


Subject(s)
Gastrointestinal Hormones/biosynthesis , Proto-Oncogene Proteins B-raf/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Adult , Aged , Carcinoma , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Line , Cell Line, Tumor , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neuropeptides/biosynthesis , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Up-Regulation
9.
Anticancer Res ; 30(10): 3981-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036711

ABSTRACT

BACKGROUND: Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic molecule restricted to endocrine glands and, particularly, to steroid-secreting cells. The expression of EG-VEGF and its significance in human adenohypophysis in physiological and pathological conditions is still unknown. MATERIALS AND METHODS: In this study, we investigated by immunohistochemistry the expression of EG-VEGF in 2 samples of normal adenohypophysis and 43 bioptic samples of pituitary adenoma. Moreover, the expression of growth hormone (GH), prolactin (PRL), follicle-stimulating hormone (FSH), luteinizing hormone (LH), thyroid-stimulating hormone (TSH) and adrenocorticoprophic hormone (ACTH) were also estimated. RESULTS: The results of this study for the first time demonstrate a down-regulation of EG-VEGF expression in human pituitary adenoma as compared to normal adenohypophysis, suggesting an impaired function of the neoplastic cells in terms of hormone release in the blood stream, as a consequence of impaired tumor angiogenesis in the tumor. CONCLUSION: On the basis of our data showing a marked decrease in the expression of EG-VEGF in pituitary adenoma, with the exception of LH-secreting adenomas, we suggest that LH might be involved in the induction of EG-VEGF secretion.


Subject(s)
Adenoma/metabolism , Pituitary Neoplasms/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Adrenocorticotropic Hormone/biosynthesis , Down-Regulation , Follicle Stimulating Hormone/biosynthesis , Human Growth Hormone/biosynthesis , Humans , Immunohistochemistry , Luteinizing Hormone/biosynthesis , Pituitary Gland, Anterior/metabolism , Prolactin/biosynthesis , Thyrotropin/biosynthesis
10.
Fertil Steril ; 93(4): 1052-60, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19135668

ABSTRACT

OBJECTIVE: To study the expression of vascular endothelial growth factor (VEGF), endocrine gland-derived VEGF (EG-VEGF/PK1), and its receptors (PKR1 and PKR2) in eutopic and ectopic endometrial tissues. DESIGN: A case-control study. SETTING: University reproduction unit. PATIENT(S): Infertile women undergoing diagnostic laparoscopy for tubal patency. INTERVENTION(S): Endometrial and endometriotic tissue sampling from women with and without endometriosis. MAIN OUTCOME MEASURE(S): Quantitative polymerase chain reaction (PCR) analysis of genes in eutopic and ectopic endometrial tissues. The EG-VEGF protein was studied by immunohistochemistry. RESULT(S): In normal endometrium, EG-VEGF messenger RNA (mRNA) expression was 50-fold higher in the secretory than in the proliferative phase, but that of PKR1 was 6-fold higher in the latter than in the former. The PKR2 transcript was detected in the proliferative but not the secretory endometrium. In patients with endometriosis, eutopic endometrial PKR2 transcript level was 4-fold higher in the proliferative than in the secretory phase. No differences in EG-VEGF or PKR1 were found in proliferative versus secretory endometrium in these patients. There were no significant differences in the expression of EG-VEGF in eutopic endometrium of normal women and in those with endometriosis. In the paired laser-captured microdissected eutopic endometrial and ectopic endometriotic samples, a significantly higher EG-VEGF, but not VEGF, transcript level was detected in the ectopic when compared with eutopic samples; whereas the expressions of PKR1 and PKR2 were barely detectable. The H-scoring confirmed that the stroma of endometriotic samples had a significantly higher EG-VEGF protein expression than that in the paired eutopic endometrium. CONCLUSION(S): High levels of EG-VEGF expression may play an important role in angiogenesis in endometriotic tissues.


Subject(s)
Choristoma/metabolism , Endocrine Glands , Endometriosis/metabolism , Endometrium , Up-Regulation/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Vascular Endothelial Growth Factors/biosynthesis , Adult , Case-Control Studies , Choristoma/pathology , Endometriosis/pathology , Female , Humans , Young Adult
11.
Pancreatology ; 9(1-2): 165-72, 2009.
Article in English | MEDLINE | ID: mdl-19077468

ABSTRACT

AIMS: Endocrine gland-derived vascular endothelial growth factor (EG-VEGF)/prokineticins have been identified as tissue-specific angiogenic factors. This study investigates the expression and localization of EG-VEGF and its receptors in pancreatic tissues and pancreatic stellate cells (PSCs). METHODS: mRNA levels of EG-VEGF/prokineticin 1 (PK1), prokineticin 2 (PK2) and their receptors 1 (PKR1) and 2 (PKR2) were measured in pancreatic tissues, pancreatic cancer cell lines and PSCs by quantitative reverse-transcriptase polymerase chain reaction (QRT-PCR). Protein expression of PK1, PKR1 and PKR2 was assessed in pancreatic tissues by immunohistochemistry. Growth factor-induced secretion of EG-VEGF was measured by ELISA. RESULTS: QRT-PCR analysis in bulk tissues of normal pancreas, chronic pancreatitis and pancreatic ductal adenocarcinoma showed no significant difference of PK1 mRNA levels, whereas PK2 mRNA was barely detectable. High PK1 mRNA levels were observed only in cultured PSCs and microdissected islet cells, but not in cancer cells, and PK1 protein was localized mainly in islets and cancer-associated stromal cells. PKR1 and PKR2 proteins were present in endothelial cells of small blood vessels. TGF-beta(1) and PDGF-BB specifically stimulated PK1 secretion in PSCs. CONCLUSIONS: Islet and/or PSC-derived PK1 might act through its receptors on endothelial cells to increase angiogenesis in pancreatic diseases.


Subject(s)
Islets of Langerhans/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Cell Line, Tumor , Cells, Cultured , Gastrointestinal Hormones/biosynthesis , Humans , Neuropeptides/biosynthesis , Pancreas/cytology , Pancreatitis, Chronic/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Peptide/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
12.
Placenta ; 28(10): 1049-58, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17531315

ABSTRACT

Compelling evidence indicates that vascular endothelial growth factor (VEGF) is an important mediator of placental angiogenesis and appears to be disregulated in pre-eclampsia (PE). Recently, we characterised the expression of EG-VEGF (endocrine gland-derived vascular endothelial growth factor), also known as prokineticin 1 (PK1) in human placenta during the first trimester of pregnancy and showed that this factor is likely to play an important role in human placentation. However, because it is impossible to prospectively study placentation in humans, it has been impossible to further characterise EG-VEGF expression throughout complete gestation and especially at critical gestational ages for PE development. In the present study, we used mouse placenta to further characterise EG-VEGF expression throughout gestation. We investigated the pattern of expression of EG-VEGF and its receptors, PKR1 and PKR2 at the mRNA and protein levels. Our results show that EG-VEGF and VEGF exhibit different patterns of expression and different localisations in the mouse placenta. EG-VEGF was mainly localised in the labyrinth whereas VEGF was mainly present in glycogen and giant cells. EG-VEGF mRNA and protein levels were highest before 10.5days post coitus (dpc) whereas those of VEGF showed stable expression throughout gestation. PKR1 protein was localised to the labyrinth layer and showed the same pattern of expression as EG-VEGF whereas PKR2 expression was maintained over 10.5dpc with both trophoblastic and endothelial cell localisations. Altogether these findings suggest that EG-VEGF may have a direct effect on both endothelial and trophoblastic cells and is likely to play an important role in mouse placentation.


Subject(s)
Placenta/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Animals , Female , Gastrointestinal Hormones/biosynthesis , Gestational Age , Immunohistochemistry , Mice , Neuropeptides/biosynthesis , Pregnancy , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
13.
Biol Reprod ; 76(5): 749-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17229935

ABSTRACT

Prokineticin 1 (PROK1), also termed endocrine gland-derived vascular endothelial growth factor (endocrine gland-derived VEGF), is a newly identified protein assigned with diverse biologic functions. It binds two homologous G protein-coupled receptors, PROKR1 and PROKR2. To better understand the roles of PROK1 and its receptors in ovarian function, their expression was determined in follicles and corpora lutea (CLs) at different developmental stages. PROK1 mRNA levels were low at early luteal stage and midluteal stage, but increased sharply during natural or induced luteolysis. High PROK1 mRNA levels also were found in atretic follicles. This profile of PROK1 expression was opposite to that of the well-established angiogenic factor VEGF. Of the two receptor-type expressions, PROKR1 but not PROKR2 was correlated positively with its ligand. Immunohistochemical staining revealed that PROK1 was located mainly within the muscular layer of arterioles, and during regression it also was localized to macrophages and steroidogenic cells. The expression pattern of ITGB2 mRNA, a leukocyte cell marker, overlapped that of PROK1, thus suggesting that leukocyte infiltration may explain the elevated expression of PROK1 in atretic follicles and regressing CL. Indeed, flow cytometry analyses showed that nearly all beta-2 integrin chain (ITGB2)-positive cells also were stained with anti-PROK1 and that significantly more ITGB2/PROK1 double-stained cells were present in degenerating follicles and CL. Furthermore, when challenged in vitro with PROK1, adherent, mononuclear cell numbers and TNF levels were elevated, indicating that PROK1 triggers monocyte activation. Together, these data suggest that PROK1, acting via PROKR1, may be involved in the recruitment of monocytes to regressing CL and atretic follicles and their consequent activation therein.


Subject(s)
Corpus Luteum/growth & development , Estrous Cycle/metabolism , Follicular Atresia/physiology , Ovary/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Animals , Cattle , Cell Count , Ceramides/metabolism , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/genetics , Dinoprost/biosynthesis , Dinoprost/genetics , Escherichia coli/metabolism , Female , Flow Cytometry , Immunohistochemistry , Monocytes/drug effects , Monocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
14.
J Comp Neurol ; 498(6): 796-809, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16927269

ABSTRACT

Prokineticins are a pair of regulatory peptides that have been shown to play important roles in gastrointestinal motility, angiogenesis, circadian rhythms, and, recently, olfactory bulb neurogenesis. Prokineticins exert their functions via activation of two closely related G-protein-coupled receptors. Here we report a comprehensive mRNA distribution for both prokineticins (PK1 and PK2) and their receptors (PKR1 and PKR2) in the adult mouse brain with the use of in situ hybridization. PK2 mRNA is expressed in discrete regions of the brain, including suprachiasmatic nucleus, islands of Calleja and medial preoptic area, olfactory bulb, nucleus accumbens shell, hypothalamic arcuate nucleus, and amygdala. PK1 mRNA is expressed exclusively in the brainstem, with high abundance in the nucleus tractus solitarius. PKR2 mRNA is detected throughout the brain, with prominent expression in olfactory regions, cortex, thalamus and hypothalamus, septum and hippocampus, habenula, amygdala, nucleus tractus solitarius, and circumventricular organs such as subfornical organ, median eminence, and area postrema. PKR2 mRNA is also detected in mammillary nuclei, periaqueductal gray, and dorsal raphe. In contrast, PKR1 mRNA is found in fewer brain regions, with moderate expression in the olfactory regions, dentate gyrus, zona incerta, and dorsal motor vagal nucleus. Both PKR1 and PKR2 are also detected in olfactory ventricle and subventricular zone of the lateral ventricle, both of which are rich sources of neuronal precursors. These extensive expression patterns suggest that prokineticins may have a broad array of functions in the central nervous system, including circadian rhythm, neurogenesis, ingestive behavior, reproduction, and autonomic function.


Subject(s)
Brain/metabolism , Gastrointestinal Hormones/biosynthesis , Neuropeptides/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Peptide/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Animals , Gene Expression , Image Processing, Computer-Assisted , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
15.
J Biol Rhythms ; 20(3): 206-18, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15851527

ABSTRACT

Prokineticin 2 (PK2) is a putative output molecule from the SCN. PK2 RNA levels are rhythmic in the mouse SCN, with high levels during the day, and PK2 administration suppresses nocturnal locomotor activity in rats. The authors examined the PK2 system in a diurnal rodent, Arvicanthis niloticus, to determine whether PK2 or PK2 receptors differ between diurnal and nocturnal species. The major transcript variant of A. niloticus PK2 (AnPK2) encodes a 26-residue signal peptide followed by the presumed mature peptide of 81 residues. Within the grass rat signal sequence, polymorphic sequences and amino acid substitutions were observed relative to mouse and laboratory rats, but the hydrophobic core and cleavage site of the signal sequence were preserved. The mature PK2 peptide is identical among A. niloticus, rat, and mouse. AnPK2 mRNA is rhythmically expressed in the SCN, with peak RNA levels occurring in the morning, preceding peaks of Per1 and Per2 as in mouse SCN. Analysis of prokineticin receptor 2 (PKR2) sequences revealed polymorphisms among the grass rats studied. PKR2 mRNA was expressed in the SCN and paraventricular nuclei of the thalamus and hypothalamus. While further analysis is necessary, there is no clear evidence indicating that a difference in the PK2 ligand/receptor system accounts for diurnality in this rodent species. These data contribute to a growing body of evidence suggesting that the key to diurnality lies downstream of the SCN in A. niloticus.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Amino Acid Sequence , Animals , Brain Chemistry/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , In Situ Hybridization , Light , Male , Molecular Sequence Data , Polymorphism, Genetic/genetics , Rats , Rodentia , Transcription, Genetic , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
16.
Br J Pharmacol ; 142(1): 181-91, 2004 May.
Article in English | MEDLINE | ID: mdl-15066905

ABSTRACT

1. The small protein Bv8, secreted by the skin of the frog Bombina variegata, belongs to a novel family of secreted proteins whose mammalian orthologues have been identified and named prokineticins (PK-1 and PK-2). 2. Bv8 (from 2.5 to 60 pmol) injected into the lateral ventricles of rat brain suppressed diurnal, nocturnal, deprivation-induced and neuropeptide Y-stimulated feeding and stimulated diurnal drinking. Nocturnal drinking was increased only in fasted rats. 3. PK-2 mRNA is expressed in discrete areas of the rat brain, including the suprachiasmatic nucleus (SCN), medial preoptic area (MPA) and nucleus of the solitary tract (NTS). In the SCN neurons, PK-2 mRNA is highest during the light phase of the circadian cycle and undetectable during the dark phase. 4. The G-protein-coupled receptor prokineticin receptor 2 (PKR-2), which binds Bv8 and PK-2 with high affinity, is mainly expressed in the piriform cortex, paraventricular thalamic nucleus, parataenial nucleus (PT), SCN, hypothalamic paraventricular (PVH) and dorsomedial (DMH) nuclei, arcuate nucleus (ARC) and subfornical organ (SFO) of the rat brain. 5. Bv8 microinjected into the ARC, at doses from 0.02 to 2.0 pmol during night-time or from 0.2 to 5 pmol in 24-h-fasted rats, selectively suppressed feeding without affecting drinking. When injected into the SFO, Bv8 (from 0.2 to 2 pmol) stimulated drinking but did not affect feeding. Bv8 injections into other brain areas left rat ingestive behaviours unchanged. 6. We hypothesize that PK-2-rich projections from SCN neurons to PKR-expressing ARC neurons could transmit the circadian rhythm of feeding, whereas inputs from the PK-2-expressing NTS neurons to the PKR-2-expressing SFO neurons could transmit visceral information on the water-electrolyte balance and osmotic regulation.


Subject(s)
Amphibian Proteins/pharmacology , Feeding Behavior/drug effects , Gastrointestinal Hormones/physiology , Neuropeptides/pharmacology , Neuropeptides/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Amphibian Proteins/administration & dosage , Animals , Anura , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drinking/drug effects , Drinking/physiology , Eating/drug effects , Eating/physiology , Feeding Behavior/physiology , Gastrointestinal Hormones/biosynthesis , Gastrointestinal Hormones/genetics , Male , Neuropeptides/administration & dosage , Neuropeptides/biosynthesis , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Structural Homology, Protein , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
17.
Biochim Biophys Acta ; 1654(1): 69-78, 2004 Mar 04.
Article in English | MEDLINE | ID: mdl-14984768

ABSTRACT

A novel family of angiogenic mitogens have been recently characterized. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF), and the mammalian homologue of Bombina variegata peptide 8 (Bv8), are two highly related endothelial cell mitogens and chemotactic factors with restricted expression profiles and selective endothelial cell activity. These peptides share two cognate G-protein coupled receptors. The expression of human EG-VEGF occurs predominantly in steroidogenic glands. Consistent with such an expression pattern, the human EG-VEGF gene promoter has a potential binding site for steroidogenic factor (SF)-1, a pivotal element for steroidogenic-specific transcription. In the human ovary, the expression of EG-VEGF is temporally and spatially complementary to the expression of VEGF-A, both in the follicular and in the luteal phase, suggesting complementary and coordinated roles of these molecules in ovarian angiogenesis. Also, EG-VEGF expression correlates with vascularity in the polycystic ovary syndrome, a leading cause of infertility. Bv8 expression is mainly restricted to the testis. The identification of these tissue-selective angiogenic factors raises the possibility that other secreted molecules with selectivity for the endothelium of other organs exist.


Subject(s)
Gastrointestinal Hormones/genetics , Neuropeptides/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Endothelium, Vascular/drug effects , Female , Follicular Phase , Gastrointestinal Hormones/biosynthesis , Gastrointestinal Hormones/pharmacology , Gene Expression Regulation , Humans , Leydig Cells/metabolism , Male , Molecular Sequence Data , Neovascularization, Pathologic/metabolism , Neuropeptides/biosynthesis , Neuropeptides/pharmacology , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , RNA, Messenger/biosynthesis , Sequence Alignment , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...