1
|
Lv P, Lu L. Diagnostic value of sFlt-1/PlGF-1 ratio and plasma PROK1 for adverse pregnancy outcomes in women with hypertensive disease of pregnancy. Kaohsiung J Med Sci 2024; 40:1068-1076. [PMID: 39625119 PMCID: PMC11618490 DOI: 10.1002/kjm2.12907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 12/06/2024] Open
Abstract
Hypertensive disease of pregnancy (HDP) is one of the most important causes of increased maternal mortality and perinatal complications during pregnancy. We investigated the pregnancy outcomes of 156 HDP patients (65 gestational hypertension [GH], 13 chronic hypertension [CH], 74 preeclampsia-eclampsia [PE-EC], and 4 superimposed on PE [CH with PE]). In patients with different types of HDP, levels of soluble fms like tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF)-1, and prokinin-1 (PROK1) were measured and compared. The predictive efficacy of these indicators was evaluated using receiving operating characteristics curves and area under the curve. Results showed that the PE cohort had a higher sFlt-1/PlGF ratio (46.12 [39.24, 68.85]) and PROK1 (498.84 [213.67, 678.30] pg/mL) level than the GH (sFlt-1/PlGF, 32.3 [21.98, 58.00], PROK1 300.77[250.0, 345.29]pg/mL) and CH cohort (sFlt-1/PlGF, 37.49 [32.68, 39.68], PROK1, 281.48 [229.25, 453.94]pg/mL). In the HDP cohort, 54 patients experienced adverse pregnancy events, and the sFlt-1/PlGF ratio, PROK1, and the combined indicators (sFlt-1/PlGF ratio and PROK1) were excellent predictors of adverse pregnancy events, especially for PE patients.
Collapse
Affiliation(s)
- Ping Lv
- Department of ObstetricalShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)Shengzhou CityZhejiang ProvinceChina
| | - Lin‐Fei Lu
- Department of ObstetricalShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)Shengzhou CityZhejiang ProvinceChina
| |
Collapse
|
2
|
Lattanzi R, Miele R. Genetic Polymorphisms of Prokineticins and Prokineticin Receptors Associated with Human Disease. Life (Basel) 2024; 14:1254. [PMID: 39459554 PMCID: PMC11509077 DOI: 10.3390/life14101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Prokineticins (PKs) are low molecular weight proteins that exert their effects by binding to two seven-transmembrane G-protein-coupled receptors (prokineticin receptors, PKRs). The prokineticin system is an important player in the development of various diseases. Several polymorphisms that are associated with infertility, neuroendocrine disorders, Hirschsprung's syndrome (HSCR), idiopathic central precocious puberty (CPP) and congenital disorders such as Kallmann syndrome (KS) have been described for both the PKs and PKR genes. The aim of this study is to summarize and describe the impact of PK/PKR polymorphisms on the pathogenesis and outcome of the above diseases, highlighting the PK system as a therapeutic target and diagnostic biomarker in pathological conditions.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
3
|
Umapathy A, Clark A, Sehgal A, Karanam V, Rajaraman G, Kalionis B, Jones H, James J, Murthi P. Molecular regulators of defective placental and cardiovascular development in fetal growth restriction. Clin Sci (Lond) 2024; 138:761-775. [PMID: 38904187 PMCID: PMC11193155 DOI: 10.1042/cs20220428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
Placental insufficiency is one of the major causes of fetal growth restriction (FGR), a significant pregnancy disorder in which the fetus fails to achieve its full growth potential in utero. As well as the acute consequences of being born too small, affected offspring are at increased risk of cardiovascular disease, diabetes and other chronic diseases in later life. The placenta and heart develop concurrently, therefore placental maldevelopment and function in FGR may have profound effect on the growth and differentiation of many organ systems, including the heart. Hence, understanding the key molecular players that are synergistically linked in the development of the placenta and heart is critical. This review highlights the key growth factors, angiogenic molecules and transcription factors that are common causes of defective placental and cardiovascular development.
Collapse
Affiliation(s)
- Anandita Umapathy
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Alys Clark
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia and Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Vijaya Karanam
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
| | - Gayathri Rajaraman
- First year college, Victoria University, St Albans, Victoria 3021, Australia
| | - Bill Kalionis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
| | - Helen N. Jones
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, U.S.A
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, U.S.A
| | - Jo James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Padma Murthi
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
4
|
Jovanovic N, Mustieles V, Althuser M, Lyon-Caen S, Alfaidy N, Thomsen C, Sakhi AK, Sabaredzovic A, Bayat S, Couturier-Tarrade A, Slama R, Philippat C. Associations between synthetic phenols, phthalates, and placental growth/function: a longitudinal cohort with exposure assessment in early pregnancy. Hum Reprod Open 2024; 2024:hoae018. [PMID: 38689737 PMCID: PMC11057944 DOI: 10.1093/hropen/hoae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/26/2024] [Indexed: 05/02/2024] Open
Abstract
STUDY QUESTION Is exposure to environmental chemicals associated with modifications of placental morphology and function? SUMMARY ANSWER Phthalates, a class of ubiquitous chemicals, showed an association with altered placental weight, placental vascular resistance (PVR), and placental efficiency. WHAT IS KNOWN ALREADY Only a few epidemiological studies have assessed the effects of phenols and phthalates on placental health. Their results were affected by exposure measurement errors linked to the rapid excretion of these compounds and the reliance on a limited number of spot urine samples to assess exposure. STUDY DESIGN SIZE DURATION A prospective mother-child cohort, with improved exposure assessment for non-persistent chemicals, recruited participants between 2014 and 2017. Sample size ranged between 355 (placental parameters measured at birth: placental weight and placental-to-fetal weight ratio (PFR): a proxy for placental efficiency) and 426 (placental parameters measured during pregnancy: placental thickness and vascular resistance). PARTICIPANTS/MATERIALS SETTING METHODS Phenols (four parabens, two bisphenols, triclosan, and benzophenone-3), 13 phthalate metabolites, and two non-phthalate plasticizer metabolites were measured in within-subject pools of repeated urine samples collected during the second and third trimesters of pregnancy (median = 21 samples/trimester/woman). Placental thickness and PVR were measured during pregnancy. The placenta was weighed at birth and the PFR was computed. Both adjusted linear regression and Bayesian Kernel Machine Regression were used to evaluate associations between phenols and phthalates (alone or as a mixture) and placental parameters. Effect modification by child sex was also investigated. MAIN RESULTS AND THE ROLE OF CHANCE Several phthalate metabolites were negatively associated with placental outcomes. Monobenzyl phthalate (MBzP) concentrations, during the second and third trimesters of pregnancy, were associated with a decrease in both placental weight at birth (β = -20.1 g [95% CI: -37.8; -2.5] and β = -17.4 g [95% CI: -33.2; -1.6], for second and third trimester, respectively) and PFR (β = -0.5 [95% CI: -1, -0.1] and β = -0.5 [95% CI: -0.9, -0.1], for the second and third trimester, respectively). Additionally, MBzP was negatively associated with PVR during the third trimester (β= -0.9 [95% CI: -1.8; 0.1]). Mono-n-butyl phthalate (MnBP), was negatively associated with PVR in both trimesters (β = -1.3, 95% CI: [-2.3, -0.2], and β = -1.2, 95% CI: [-2.4, -0.03], for the second and third trimester, respectively). After stratification for child sex, Σ diisononyl phthalate (DiNP) (either second or third-trimester exposures, depending on the outcomes considered) was associated with decreased PVR in the third trimester, as well as decreased placental weight and PFR in males. No associations were observed for phenol biomarkers. LIMITATIONS REASONS FOR CAUTION False positives cannot be ruled out. Therefore, chemicals that were associated with multiple outcomes (MnBP and DiNP) or reported in existing literature as associated with placental outcomes (MBzP) should be considered as the main results. WIDER IMPLICATIONS OF THE FINDINGS Our results are consistent with in vitro studies showing that phthalates target peroxisome proliferator-activated receptor γ, in the family of nuclear receptors involved in key placental development processes such as trophoblast proliferation, migration, and invasion. In addition to placental weight at birth, we studied placental parameters during pregnancy, which could provide a broader view of how environmental chemicals affect maternal-fetal exchanges over the course of pregnancy. Our findings contribute to the increasing evidence indicating adverse impacts of phthalate exposure on placental health. STUDY FUNDING/COMPETING INTERESTS This work was supported by the French Research Agency-ANR (MEMORI project ANR-21-CE34-0022). The SEPAGES cohort was supported by the European Research Council (N°311765-E-DOHaD), the European Community's Seventh Framework Programme (FP7/2007-206-N°308333-892 HELIX), the European Union's Horizon 2020 research and innovation programme (N° 874583 ATHLETE Project, N°825712 OBERON Project), the French Research Agency-ANR (PAPER project ANR-12-PDOC-0029-01, SHALCOH project ANR-14-CE21-0007, ANR-15-IDEX-02 and ANR-15-IDEX5, GUMME project ANR-18-CE36-005, ETAPE project ANR-18-CE36-0005-EDeN project ANR-19-CE36-0003-01), the French Agency for Food, Environmental and Occupational Health & Safety-ANSES (CNAP project EST-2016-121, PENDORE project EST-2016-121, HyPAxE project EST-2019/1/039, PENDALIRE project EST-2022-169), the Plan Cancer (Canc'Air project), the French Cancer Research Foundation Association de Recherche sur le Cancer-ARC, the French Endowment Fund AGIR for chronic diseases-APMC (projects PRENAPAR, LCI-FOT, DysCard), the French Endowment Fund for Respiratory Health, the French Fund-Fondation de France (CLIMATHES-00081169, SEPAGES 5-00099903, ELEMENTUM-00124527). N.J. was supported by a doctoral fellowship from the University Grenoble Alpes. V.M. was supported by a Sara Borrell postdoctoral research contract (CD22/00176), granted by Instituto de Salud Carlos III (Spain) and NextGenerationEU funds. The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT02852499.
Collapse
Affiliation(s)
- Nicolas Jovanovic
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Vicente Mustieles
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
- Department of Radiology and Physical Medicine, University of Granada, Biomedical Research Center (CIBM), Granada, Spain
| | - Marc Althuser
- Department of Obstetrics/Gynecology and Fetal Medicine, Grenoble University Hospital, Grenoble, France
| | - Sarah Lyon-Caen
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Nadia Alfaidy
- Commissariat à l'Energie Atomique (CEA), IRIG department, INSERM U1292, and Grenoble Alpes University (UGA), Grenoble, France
| | | | | | | | - Sam Bayat
- Department of Obstetrics/Gynecology and Fetal Medicine, Grenoble University Hospital, Grenoble, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Rémy Slama
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Claire Philippat
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
5
|
Goto S, Ozaki Y, Ozawa F, Yoshihara H, Ujvari D, Kitaori T, Sugiura-Ogasawara M. Impaired decidualization and relative increase of PROK1 expression in the decidua of patients with unexplained recurrent pregnancy loss showing insulin resistance. J Reprod Immunol 2023; 160:104155. [PMID: 37801889 DOI: 10.1016/j.jri.2023.104155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
A recent meta-analysis revealed that patients with unexplained recurrent pregnancy loss (RPL) show higher insulin resistance compared to healthy controls. However, the etiology of RPL remains unknown. Prokineticin (PROK1), a pleiotropic uterine endometrial protein, is important for implantation and decidualization and is regulated by hypoxia and insulin. In this study, we investigated the decidualization status and the role of PROK1 in the decidua of patients with unexplained RPL showing insulin resistance. Thirty-two patients with unexplained RPL were included in this study. Following the diagnosis of a miscarriage, the decidua and villi of the patient were surgically collected. Fasting blood glucose and insulin levels were measured, and HOMA-β was calculated. Using IHC and ELISA, the expression of IGFBP-1, PRL and PROK1 in the decidua and IGF-2 in the villi were analyzed in patients with euploid miscarriage with a high HOMA-β index (n = 8) and compared to controls (euploid miscarriage with normal HOMA-β: n = 12, aneuploid miscarriage with normal HOMA-β: n = 12). The co-localization of PROK1 and IGFBP-1 was observed in the decidua by IHC. In the decidua of RPL patients with high HOMA-β, the expression levels of IGFBP-1 and PRL were significantly lower, whereas the PROK1/IGFBP-1 ratio was significantly higher compared to that of the controls. IGF-2 expression in villi was significantly lower in RPL patients with high HOMA-β. Impaired decidualization and excessive PROK1 production may have pathological implications in patients with unexplained RPL with insulin resistance, especially under the state of hyper insulin production.
Collapse
Affiliation(s)
- Shinobu Goto
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan.
| | - Yasuhiko Ozaki
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan; Department of Obstetrics and Gynecology, Nagoya City University West Medical Center, 1-1-1 Hirate-cho, Kita-ku, Nagoya, Aichi ZIP 462-8508, Japan
| | - Fumiko Ozawa
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| | - Hiroyuki Yoshihara
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan; Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institute, Solna, Sweden
| | - Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden; Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Tamao Kitaori
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| | - Mayumi Sugiura-Ogasawara
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| |
Collapse
|
6
|
Ujvari D, Trouva A, Hirschberg AL, Vanky E. Maternal serum levels of prokineticin-1 related to pregnancy complications and metformin use in women with polycystic ovary syndrome: a post hoc analysis of two prospective, randomised, placebo-controlled trials. BMJ Open 2023; 13:e073619. [PMID: 37989369 PMCID: PMC10668301 DOI: 10.1136/bmjopen-2023-073619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/26/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVE Serum prokineticin-1 (s-PROK1) in the second and third trimester of pregnancy is positively correlated to preeclampsia, intrauterine growth restriction (IUGR) and preterm delivery. Women with polycystic ovary syndrome (PCOS) are prone to these adverse pregnancy outcomes. However, the contribution of PROK1 to the development of pregnancy complications and the effect of metformin and hyperandrogenism on s-PROK1 in PCOS have not been studied previously. DESIGN This work is a post hoc analysis of two prospective, randomised, placebo-controlled trials. SETTING Pregnant women with PCOS were included from 11 study centres in Norway. PARTICIPANTS From 313 women, 264 participated in the present study after exclusions due to dropouts or insufficient serum samples. INTERVENTION Women with PCOS were randomly administered with metformin or placebo, from first trimester to delivery. PRIMARY AND SECONDARY OUTCOME MEASURES s-PROK1 was analysed using ELISA at gestational week 19 and related to pregnancy complications, fasting insulin levels, homoeostatic model assessment for insulin resistance (HOMA-IR), testosterone, or androstenedione levels, metformin use, PCOS phenotype and hyperandrogenism. RESULTS Maternal s-PROK1 in the second trimester did not predict pregnancy-induced hypertension, pre-eclampsia or late miscarriage/preterm delivery in women with PCOS. However, s-PROK1 was lower in women who used metformin before inclusion, both in those randomised to metformin and to placebo, compared with those who did not. s-PROK1 was also lower in those who used metformin both at conception and during pregnancy compared with those who used metformin from inclusion or did not use metformin at all. s-PROK1 was lower in hyperandrogenic compared with normo-androgenic women with PCOS. CONCLUSIONS Maternal s-PROK1 in the second trimester did not predict pregnancy complications in PCOS. Those who used metformin at conception and/or during pregnancy had lower s-PROK1. PCOS women with hyperandrogenism exhibited lower s-PROK1 compared with normo-adrogenic phenotypes. TRIAL REGISTRATION NUMBER NCT03259919 and NCT00159536.
Collapse
Affiliation(s)
- Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology; National Pandemic Centre; Centre for Translational Microbiome Research, Karolinska Institute, Solna, Sweden
| | - Anastasia Trouva
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Gynaecology and Obstetrics, St Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
7
|
Raia-Barjat T, Chauleur C, Collet C, Rancon F, Hoffmann P, Desseux M, Lemaitre N, Benharouga M, Giraud A, Alfaidy N. EG-VEGF maternal levels predict spontaneous preterm birth in the second and third trimesters in pregnant women with risk factors for placenta-mediated complications. Sci Rep 2023; 13:19921. [PMID: 37963927 PMCID: PMC10645734 DOI: 10.1038/s41598-023-46883-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023] Open
Abstract
Prediction of spontaneous preterm birth in asymptomatic women remains a great challenge for the public health system. The aim of the study was to determine the informational value of EG-VEGF circulating levels for prediction of spontaneous preterm birth in the second and third trimesters in pregnant women at high risk for placenta-mediated complications. A prospective multicenter cohort study including 200 pregnant patients with five-serum sampling per patient. Women with spontaneous preterm birth have higher concentrations of serum EG-VEGF than uncomplicated patients at 24 weeks, 28 weeks and 32 weeks (p = 0.03, 0.02 and < 0.001). The areas under the curve reached 0.9 with 100% sensitivity at 32 weeks for the prediction of spontaneous preterm birth. Serum EG-VEGF concentrations could be considered as a reliable biomarker of spontaneous preterm birth in high-risk for placenta-mediated complications pregnant women.
Collapse
Affiliation(s)
- Tiphaine Raia-Barjat
- Department of Gynecology and Obstetrics, Hôpital Nord, University Hospital, Centre Hospitalier Universitaire de Saint-Étienne, Avenue Albert Raimond, 42270, Saint-Étienne, Saint Priest en Jarez, France.
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France.
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France.
| | - Céline Chauleur
- Department of Gynecology and Obstetrics, Hôpital Nord, University Hospital, Centre Hospitalier Universitaire de Saint-Étienne, Avenue Albert Raimond, 42270, Saint-Étienne, Saint Priest en Jarez, France
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
| | - Constance Collet
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Florence Rancon
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
- INSERM, Centre d'Investigation Clinique 1408, Saint-Étienne, France
| | - Pascale Hoffmann
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Morgane Desseux
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Nicolas Lemaitre
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Mohamed Benharouga
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Antoine Giraud
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
- Neonatal Intensive Care Unit, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Nadia Alfaidy
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France.
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France.
- Université Grenoble Alpes (UGA), Grenoble, France.
| |
Collapse
|
8
|
Vincenzi M, Kremić A, Jouve A, Lattanzi R, Miele R, Benharouga M, Alfaidy N, Migrenne-Li S, Kanthasamy AG, Porcionatto M, Ferrara N, Tetko IV, Désaubry L, Nebigil CG. Therapeutic Potential of Targeting Prokineticin Receptors in Diseases. Pharmacol Rev 2023; 75:1167-1199. [PMID: 37684054 PMCID: PMC10595023 DOI: 10.1124/pharmrev.122.000801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 09/10/2023] Open
Abstract
The prokineticins (PKs) were discovered approximately 20 years ago as small peptides inducing gut contractility. Today, they are established as angiogenic, anorectic, and proinflammatory cytokines, chemokines, hormones, and neuropeptides involved in variety of physiologic and pathophysiological pathways. Their altered expression or mutations implicated in several diseases make them a potential biomarker. Their G-protein coupled receptors, PKR1 and PKR2, have divergent roles that can be therapeutic target for treatment of cardiovascular, metabolic, and neural diseases as well as pain and cancer. This article reviews and summarizes our current knowledge of PK family functions from development of heart and brain to regulation of homeostasis in health and diseases. Finally, the review summarizes the established roles of the endogenous peptides, synthetic peptides and the selective ligands of PKR1 and PKR2, and nonpeptide orthostatic and allosteric modulator of the receptors in preclinical disease models. The present review emphasizes the ambiguous aspects and gaps in our knowledge of functions of PKR ligands and elucidates future perspectives for PK research. SIGNIFICANCE STATEMENT: This review provides an in-depth view of the prokineticin family and PK receptors that can be active without their endogenous ligand and exhibits "constitutive" activity in diseases. Their non- peptide ligands display promising effects in several preclinical disease models. PKs can be the diagnostic biomarker of several diseases. A thorough understanding of the role of prokineticin family and their receptor types in health and diseases is critical to develop novel therapeutic strategies with safety concerns.
Collapse
Affiliation(s)
- Martina Vincenzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Amin Kremić
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Appoline Jouve
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Roberta Lattanzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Rossella Miele
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Mohamed Benharouga
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Nadia Alfaidy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Stephanie Migrenne-Li
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Anumantha G Kanthasamy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Marimelia Porcionatto
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Napoleone Ferrara
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Igor V Tetko
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Laurent Désaubry
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Canan G Nebigil
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| |
Collapse
|
9
|
Younes H, Kyritsi I, Mahrougui Z, Benharouga M, Alfaidy N, Marquette C. Effects of Prokineticins on Cerebral Cell Function and Blood-Brain Barrier Permeability. Int J Mol Sci 2023; 24:15428. [PMID: 37895111 PMCID: PMC10607385 DOI: 10.3390/ijms242015428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Prokineticins are a family of small proteins with diverse roles in various tissues, including the brain. However, their specific effects on different cerebral cell types and blood-brain barrier (BBB) function remain unclear. The aim of this study was to investigate the effects of PROK1 and PROK2 on murine cerebral cell lines, bEnd.3, C8.D30, and N2a, corresponding to microvascular endothelial cells, astrocytes and neurons, respectively, and on an established BBB co-culture model. Western blot analysis showed that prokineticin receptors (PROKR1 and PROKR2) were differentially expressed in the considered cell lines. The effect of PROK1 and PROK2 on cell proliferation and migration were assessed using time-lapse microscopy. PROK1 decreased neural cells' proliferation, while it had no effect on the proliferation of endothelial cells and astrocytes. In contrast, PROK2 reduced the proliferation of all cell lines tested. Both PROK1 and PROK2 increased the migration of all cell lines. Blocking PROKRs with the PROKR1 antagonist (PC7) and the PROKR2 antagonist (PKR-A) inhibited astrocyte PROK2-mediated migration. Using the insert co-culture model of BBB, we demonstrated that PROKs increased BBB permeability, which could be prevented by PROKRs' antagonists.
Collapse
Affiliation(s)
- Hadi Younes
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Ioanna Kyritsi
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Zineb Mahrougui
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Mohamed Benharouga
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Nadia Alfaidy
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Christel Marquette
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| |
Collapse
|
10
|
Zhao X, Zhang Z, Zhu Q, Luo Y, Ye Q, Shi S, He X, Zhu J, Zhang D, Xia W, Zhang Y, Jiang L, Cui L, Ye Y, Xiang Y, Hu J, Zhang J, Lin CP. Modeling human ectopic pregnancies with trophoblast and vascular organoids. Cell Rep 2023; 42:112546. [PMID: 37224015 DOI: 10.1016/j.celrep.2023.112546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/15/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Ruptured ectopic pregnancy (REP), a pregnancy complication caused by aberrant implantation, deep invasion, and overgrowth of embryos in fallopian tubes, could lead to rupture of fallopian tubes and accounts for 4%-10% of pregnancy-related deaths. The lack of ectopic pregnancy phenotypes in rodents hampers our understanding of its pathological mechanisms. Here, we employed cell culture and organoid models to investigate the crosstalk between human trophoblast development and intravillous vascularization in the REP condition. Compared with abortive ectopic pregnancy (AEP), the size of REP placental villi and the depth of trophoblast invasion are correlated with the extent of intravillous vascularization. We identified a key pro-angiogenic factor secreted by trophoblasts, WNT2B, that promotes villous vasculogenesis, angiogenesis, and vascular network expansion in the REP condition. Our results reveal the important role of WNT-mediated angiogenesis and an organoid co-culture model for investigating intricate communications between trophoblasts and endothelial/endothelial progenitor cells.
Collapse
Affiliation(s)
- Xiaoya Zhao
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Zhenwu Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qian Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Yurui Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shuxiang Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xueyang He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Jing Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Duo Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Wei Xia
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Yiqin Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Long Cui
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yinghui Ye
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Jian Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Road, Shanghai 200030, China; Shanghai Municipal Key Clinical Specialty, Shanghai, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
11
|
Baryla M, Goryszewska-Szczurek E, Kaczynski P, Balboni G, Waclawik A. Prokineticin 1 is a novel factor regulating porcine corpus luteum function. Sci Rep 2023; 13:5085. [PMID: 36991037 PMCID: PMC10060428 DOI: 10.1038/s41598-023-32132-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Monika Baryla
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Goryszewska-Szczurek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Piotr Kaczynski
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Gianfranco Balboni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042, Monserrato, Cagliari, Italy
| | - Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
12
|
Hu Y, Qi S, Zhuang H, Zhuo Q, Liang Y, Kong H, Zhao C, Zhang S. Proteotranscriptomic analyses reveal distinct interferon-beta signaling pathways and therapeutic targets in choroidal neovascularization. Front Immunol 2023; 14:1163739. [PMID: 37025993 PMCID: PMC10071000 DOI: 10.3389/fimmu.2023.1163739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 04/08/2023] Open
Abstract
Aim To investigate the molecular mechanism underlying the onset of choroidal neovascularization (CNV). Methods Integrated transcriptomic and proteomic analyses of retinas in mice with laser-induced CNV were performed using RNA sequencing and tandem mass tag. In addition, the laser-treated mice received systemic interferon-β (IFN-β) therapy. Measurements of CNV lesions were acquired by the confocal analysis of stained choroidal flat mounts. The proportions of T helper 17 (Th17) cells were determined by flow cytometric analysis. Results A total of differentially expressed 186 genes (120 up-regulated and 66 down-regulated) and 104 proteins (73 up-regulated and 31 down-regulated) were identified. The gene ontology and KEGG pathway analyses indicated that CNV was mainly associated with immune and inflammatory responses, such as cellular response to IFN-β and Th17 cell differentiation. Moreover, the key nodes of the protein-protein interaction network mainly involved up-regulated proteins, including alpha A crystallin and fibroblast growth factor 2, and were verified by Western blotting. To confirm the changes in gene expression, real-time quantitative PCR was performed. Furthermore, levels of IFN-β in both the retina and plasma, as measured by enzyme-linked immunosorbent assay (ELISA), were significantly lower in the CNV group than in the control group. IFN-β treatment significantly reduced CNV lesion size and promoted the proliferation of Th17 cells in laser-treated mice. Conclusions This study demonstrates that the occurrence of CNV might be associated with the dysfunction of immune and inflammatory processes and that IFN-β could serve as a potential therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chen Zhao
- *Correspondence: Chen Zhao, ; Shujie Zhang,
| | | |
Collapse
|
13
|
Huang KH, Chen FY, Liu ZZ, Luo JY, Xu RL, Jiang LL, Yan JY. Prediction of pre-eclampsia complicated by fetal growth restriction and its perinatal outcome based on an artificial neural network model. Front Physiol 2022; 13:992040. [DOI: 10.3389/fphys.2022.992040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022] Open
Abstract
Objective: Pre-eclampsia (PE) complicated by fetal growth restriction (FGR) increases both perinatal mortality and the incidence of preterm birth and neonatal asphyxia. Because ultrasound measurements are bone markers, soft tissues, such as fetal fat and muscle, are ignored, and the selection of section surface and the influence of fetal position can lead to estimation errors. The early detection of FGR is not easy, resulting in a relative delay in intervention. It is assumed that FGR complicated with PE can be predicted by laboratory and clinical indicators. The present study adopts an artificial neural network (ANN) to assess the effect and predictive value of changes in maternal peripheral blood parameters and clinical indicators on the perinatal outcomes in patients with PE complicated by FGR.Methods: This study used a retrospective case-control approach. The correlation between maternal peripheral blood parameters and perinatal outcomes in pregnant patients with PE complicated by FGR was retrospectively analyzed, and an ANN was constructed to assess the value of the changes in maternal blood parameters in predicting the occurrence of PE complicated by FGR and adverse perinatal outcomes.Results: A total of 15 factors—maternal age, pre-pregnancy body mass index, inflammatory markers (neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio), coagulation parameters (prothrombin time and thrombin time), lipid parameters (high-density lipoprotein, low-density lipoprotein, and triglyceride counts), platelet parameters (mean platelet volume and plateletcrit), uric acid, lactate dehydrogenase, and total bile acids—were correlated with PE complicated by FGR. A total of six ANNs were constructed with the adoption of these parameters. The accuracy, sensitivity, and specificity of predicting the occurrence of the following diseases and adverse outcomes were respectively as follows: 84.3%, 97.7%, and 78% for PE complicated by FGR; 76.3%, 97.3%, and 68% for provider-initiated preterm births,; 81.9%, 97.2%, and 51% for predicting the severity of FGR; 80.3%, 92.9%, and 79% for premature rupture of membranes; 80.1%, 92.3%, and 79% for postpartum hemorrhage; and 77.6%, 92.3%, and 76% for fetal distress.Conclusion: An ANN model based on maternal peripheral blood parameters has a good predictive value for the occurrence of PE complicated by FGR and its adverse perinatal outcomes, such as the severity of FGR and preterm births in these patients.
Collapse
|
14
|
Wujcicka WI, Kacerovsky M, Krygier A, Krekora M, Kaczmarek P, Grzesiak M. Association of Single Nucleotide Polymorphisms from Angiogenesis-Related Genes, ANGPT2, TLR2 and TLR9, with Spontaneous Preterm Labor. Curr Issues Mol Biol 2022; 44:2939-2955. [PMID: 35877427 PMCID: PMC9322696 DOI: 10.3390/cimb44070203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/07/2022] Open
Abstract
In this study, we hypothesized that the changes localized at angiopoietin-2 (ANGPT2), granulocyte-macrophage colony-stimulating factor (CSF2), fms-related tyrosine kinase 1 (FLT1) and toll-like receptor (TLR) 2, TLR6 and TLR9 genes were associated with spontaneous preterm labor (PTL), as well as with possible genetic alterations on PTL-related coagulation. This case-control genetic association study aimed to identify single nucleotide polymorphisms (SNPs) for the aforementioned genes, which are correlated with genetic risk or protection against PTL in Polish women. The study was conducted in 320 patients treated between 2016 and 2020, including 160 women with PTL and 160 term controls in labor. We found that ANGPT2 rs3020221 AA homozygotes were significantly less common in PTL cases than in controls, especially after adjusting for activated partial thromboplastin time (APTT) and platelet (PLT) parameters. TC heterozygotes for TLR2 rs3804099 were associated with PTL after correcting for anemia, vaginal bleeding, and history of threatened miscarriage or PTL. TC and CC genotypes in TLR9 rs187084 were significantly less common in women with PTL, compared to the controls, after adjusting for bleeding and gestational diabetes. For the first time, it was shown that three polymorphisms-ANGPT2 rs3020221, TLR2 rs3804099 and TLR9 rs187084 -were significantly associated with PTL, adjusted by pregnancy development influencing factors.
Collapse
Affiliation(s)
- Wioletta Izabela Wujcicka
- Scientific Laboratory of the Center of Medical Laboratory Diagnostics and Screening, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
- Correspondence: or ; Tel.: +48-42-271-15-20; Fax: +48-42-271-15-10
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic;
- Biomedical Research Center, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
| | - Adrian Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Michał Krekora
- Department of Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland;
| | - Piotr Kaczmarek
- Department of Gynecology, Reproduction and Fetal Therapy, and Diagnostics and Treatment of Infertility, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
| | - Mariusz Grzesiak
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland;
- Department of Perinatology, Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| |
Collapse
|
15
|
LncRNA HOTTIP facilitates osteogenic differentiation in bone marrow mesenchymal stem cells and induces angiogenesis via interacting with TAF15 to stabilize DLX2. Exp Cell Res 2022; 417:113226. [DOI: 10.1016/j.yexcr.2022.113226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/24/2022] [Accepted: 05/22/2022] [Indexed: 12/12/2022]
|
16
|
Wasilah S, Soeatmadji DW, Sardjono TW, Kania N, Noor MS. Coal Dust Exposures Change the Spiral Artery Remodeling and Natural Killer Cells Counts in the Uterus of Pregnant Rats. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Indonesia has numerous coal resources, but the effects of environmental pollution by coal dust to human health, especially the reproductive system, are still less investigated. Chronic coal exposures during pregnancy might cause systemic and uterine inflammation that induces disturbances of spiral artery remodeling.
AIM: This research was conducted to analyze the effect of coal dust exposures to uterine spiral artery remodeling and natural killer (NK) cell counts.
METHODS: There were 42 female adult Rattus norvegicus rats which had been simultaneously mated after synchronization of estrous cycle used as animal subjects. The rats then divided into seven groups, those were K0 (without exposure); K1-1, K1-2, K1-3; and K2-1, K2-2, K2-3 which were exposed to 6.25 mg/m3, 12.5 mg/m3, and 25 mg/m3 dose for 1 h (K1) and 2 h (K2) per day, respectively. The rats were exposed by a dust exposure machine (NKBS-1-2010- 0.5) every morning starting from the day-1 to day-19 post-mating and were sacrificed on day-19 afternoon. All uterus lobes of each pregnant rat were taken and histologically processed by HE staining. Five histological slides were randomly taken as samples representing each pregnant rat and were examined for ten visual fields per slide for the measurements of diameter and wall thickness of spiral arteries. NK cells were observed immunobiological. Data analyses used Kruskal–Wallis.
RESULTS: Result showed that there were significant differences of diameters and wall thickness of spiral arteries and uterine NK cells in all exposed groups compared to control (P < 0.05). Diameter and wall thickness of spiral arteries were thicker in coal dust exposure than normal pregnant rats. NK cell number was more in coal dust exposure than normal pregnant.
CONCLUSION: Coal dust exposures caused the thickening of wall and narrowing of lumen of spiral arteries and NK cell counts that might influence pregnancy.
Collapse
|
17
|
Weng D, Han T, Dong J, Zhang M, Mi Y, He Y, Li X, Zhu X. Angiogenin and MMP-2 as potential biomarkers in the differential diagnosis of gestational trophoblastic diseases. Medicine (Baltimore) 2022; 101:e28768. [PMID: 35119039 PMCID: PMC8812619 DOI: 10.1097/md.0000000000028768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/14/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Gestational trophoblastic diseases (GTDs) are characterized by vascular abnormalities of the trophoblast, but their pathogenesis is unknown. Angiogenin (ANG) and matrix metalloproteinase (MMP)-2, which are molecules implicated in the angiogenic process, may play some role in this process. MATERIAL AND METHODS We determined ANG and MMP-2 in the placental tissues of 26 patients who had a benign mole (BM), 12 patients with gestational trophoblast neoplasia (GTN) (1 invasive hydatidiform mole, 10 choriocarcinomas, and 1 placental-site trophoblastic tumor), and 28 normal chorionic villi (NCV) subjects using immunohistochemistry staining. We obtained the serum samples from 20 patients with GTDs and 20 early pregnant women and evaluated them by the enzyme linked immunosorbent assay. RESULTS ANG expression in GTN (66.7%) and BM (100%) samples were both significantly higher (strong/intermediate staining) than in NCV (60.7%) samples (P < .001). Similarly, the immunoreactivities of MMP-2 in the GTN (66.7%) and BM (80.8%) samples were significantly elevated compared to that of the NCV (57.1%) samples (P < .001). The levels of ANG and MMP-2 in the maternal serum of the GTN group were both significantly higher than those of the control group (P < .001). ANG and MMP-2 expressions were associated with gestation age, clinical stage, and FIGO stage. A positive correlation between ANG and MMP-2 expression was observed (rs = 0.725; P < .01). CONCLUSION ANG and MMP-2 levels were significantly elevated in the placental tissues and maternal serum from patients with GTDs. Further studies with more patients may clarify the vascular abnormalities in GTDs and determine potential biomarkers in the differential diagnosis of GTDs.
Collapse
Affiliation(s)
- Dan Weng
- Department of Obstetrics and Gynecology, Hainan Hospital of PLA General Hospital, Sanya, China
- Department of Obstetrics and Gynecology, Shaanxi Provincial Maternal and Child Health's Hospital, Xi’an, China
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Tao Han
- Department of Orthopedics, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Jin Dong
- Department of Obstetrics and Gynecology, Shaanxi Provincial Maternal and Child Health's Hospital, Xi’an, China
| | - Ming Zhang
- Department of Obstetrics and Gynecology, Shaanxi Provincial Maternal and Child Health's Hospital, Xi’an, China
| | - Yang Mi
- Department of Obstetrics and Gynecology, Shaanxi Provincial Maternal and Child Health's Hospital, Xi’an, China
| | - Yiping He
- Department of Obstetrics and Gynecology, Northwestern Women's and Children's Hospital, Xi’an, China
| | - Xiaojuan Li
- Department of Obstetrics and Gynecology, Northwestern Women's and Children's Hospital, Xi’an, China
| | - Xiaoming Zhu
- Department of Obstetrics and Gynecology, Hainan Hospital of PLA General Hospital, Sanya, China
| |
Collapse
|
18
|
Gao L, Cui AQ, Wang J, Chen J, Zhang XY, Lin ZJ, Chen YH, Zhang C, Wang H, Xu DX. Paternal exposure to microcystin-LR induces fetal growth restriction partially through inhibiting cell proliferation and vascular development in placental labyrinth. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:60032-60040. [PMID: 34155591 DOI: 10.1007/s11356-021-14725-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/01/2021] [Indexed: 06/13/2023]
Abstract
Microcystin-leucine arginine (MC-LR) has reproductive and developmental toxicities. Previous studies indicated that gestational exposure to MC-LR induced fetal growth restriction in mice. The aim of this study was to further evaluate the effect of paternal MC-LR exposure before mating on fetal development. Male mice were intraperitoneally injected with either normal saline or MC-LR (10 μg/kg) daily for 35 days. Male mouse was then mated with female mice with 1:1 ratio. There was no significant difference on the rates of mating and pregnancy between MC-LR-exposed male mice and controls. Body weight and crown-rump length were reduced in fetuses whose fathers were exposed to MC-LR. Despite no difference on relative thickness of labyrinthine layer, cell proliferation, as measured by Ki67 immunostaining, was reduced in labyrinth layer of MC-LR-exposed mice. Moreover, blood sinusoid area in labyrinth layer was decreased in the fetus whose father was exposed to MC-LR before mating. Correspondingly, cross-sectional area of CD34-positive blood vessel in labyrinth layer was lower in fetuses whose fathers were exposed to MC-LR than in controls. These results provide evidence that paternal MC-LR exposure before mating induces fetal growth restriction partially through inhibiting cell proliferation and vascular development in labyrinth layer.
Collapse
Affiliation(s)
- Lan Gao
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - An-Qi Cui
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Jing Wang
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Jing Chen
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Yi Zhang
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Zhi-Jing Lin
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Yuan-Hua Chen
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Cheng Zhang
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China.
| | - De-Xiang Xu
- Department of Toxicology & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
19
|
Hirschberg AL, Jakson I, Graells Brugalla C, Salamon D, Ujvari D. Interaction between insulin and androgen signalling in decidualization, cell migration and trophoblast invasion in vitro. J Cell Mol Med 2021; 25:9523-9532. [PMID: 34463022 PMCID: PMC8505820 DOI: 10.1111/jcmm.16892] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/15/2023] Open
Abstract
Finely tuned decidualization of endometrial stromal fibroblasts into decidual cells is crucial for successful implantation and a healthy pregnancy. Both insulin and androgens are known to modulate decidualization, however, their complex effect on this process has not been fully elucidated. As hyperinsulinemia and hyperandrogenism are associated in clinical conditions, we aimed to investigate the interaction between insulin and androgens on decidualization. Primary human endometrial stromal cells were decidualized in vitro in the presence of insulin and/or androgens (dihydrotestosterone (DHT), testosterone). Gene or protein expressions of decidualization markers were measured, and cells size characteristics were determined. Migration of decidualizing endometrial stromal cells and invasion of HTR‐8/SVneo trophoblast spheroids were assessed. We found that insulin and androgens in combination enhanced the upregulation of several decidualization markers including prolactin, tissue factor, tissue inhibitor of matrix metalloproteinase 3 and connexin‐43, and also interacted in modulating cell size characteristics resulting in enlarged decidualizing cells. However, insulin and DHT together restricted the migration of decidualizing cells and invasion of trophoblast spheroids. Our findings suggest that insulin and androgens interact to potentiate the process of decidualization. On the other hand, inhibited cell migration and trophoblast invasion might negatively impact the function of decidualizing endometrial stromal cells.
Collapse
Affiliation(s)
- Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ivika Jakson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Daniel Salamon
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Molecular Changes on Maternal-Fetal Interface in Placental Abruption-A Systematic Review. Int J Mol Sci 2021; 22:ijms22126612. [PMID: 34205566 PMCID: PMC8235312 DOI: 10.3390/ijms22126612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 12/21/2022] Open
Abstract
Placental abruption is the separation of the placenta from the lining of the uterus before childbirth. It is an infrequent perinatal complication with serious after-effects and a marked risk of maternal and fetal mortality. Despite the fact that numerous placental abruption risk factors are known, the pathophysiology of this issue is multifactorial and not entirely clear. The aim of this review was to examine the current state of knowledge concerning the molecular changes on the maternal–fetal interface occurring in placental abruption. Only original research articles describing studies published in English until the 15 March 2021 were considered eligible. Reviews, book chapters, case studies, conference papers and opinions were excluded. The systematic literature search of PubMed/MEDLINE and Scopus databases identified 708 articles, 22 of which were analyzed. The available evidence indicates that the disruption of the immunological processes on the maternal–fetal interface plays a crucial role in the pathophysiology of placental abruption. The features of chronic non-infectious inflammation and augmented immunological cytotoxic response were found to be present in placental abruption samples in the reviewed studies. Various molecules participate in this process, with only a few being examined. More advanced research is needed to fully explain this complicated process.
Collapse
|
21
|
Ma H, Jiang S, Du L, Liu J, Xu X, Lu X, Ma L, Zhu H, Wei J, Yu Y. Conditioned medium from primary cytotrophoblasts, primary placenta-derived mesenchymal stem cells, or sub-cultured placental tissue promoted HUVEC angiogenesis in vitro. Stem Cell Res Ther 2021; 12:141. [PMID: 33596987 PMCID: PMC7890636 DOI: 10.1186/s13287-021-02192-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Background As a large capillary network, the human placenta plays an important role throughout pregnancy. Placental vascular development is complex and delicate and involves many types of placental cells, such as trophoblasts, and mesenchymal stem cells. There has been no systematic, comparative study on the roles of these two groups of placental cells and the whole placental tissue in the placental angiogenesis. In this study, primary cytotrophoblasts (CTBs) from early pregnancy and primary human placenta-derived mesenchymal stem cells (hPDMSCs) from different stages of pregnancy were selected as the cell research objects, and full-term placental tissue was selected as the tissue research object to detect the effects of their conditioned medium (CM) on human umbilical vein endothelial cell (HUVEC) angiogenesis. Methods We successfully isolated primary hPDMSCs and CTBs, collected CM from these placental cells and sub-cultured placental tissue, and then evaluated the effects of the CM on a series of angiogenic processes in HUVECs in vitro. Furthermore, we measured the levels of angiogenic factors in the CM of placental cells or tissue by an angiogenesis antibody array. Results The results showed that not only placental cells but also sub-cultured placental tissue, to some extent, promoted HUVEC angiogenesis in vitro by promoting proliferation, adhesion, migration, invasion, and tube formation. We also found that primary placental cells in early pregnancy, whether CTBs or hPDMSCs, played more significant roles than those in full-term pregnancy. Placental cell-derived CM collected at 24 h or 48 h had the best effect, and sub-cultured placental tissue-derived CM collected at 7 days had the best effect among all the different time points. The semiquantitative angiogenesis antibody array showed that 18 of the 43 angiogenic factors had obvious spots in placental cell-derived CM or sub-cultured placental tissue-derived CM, and the levels of 5 factors (including CXCL-5, GRO, IL-6, IL-8, and MCP-1) were the highest in sub-cultured placental tissue-derived CM. Conclusions CM obtained from placental cells (primary CTBs or hPDMSCs) or sub-cultured placental tissue contained proangiogenic factors and promoted HUVEC angiogenesis in vitro. Therefore, our research is helpful to better understand placental angiogenesis regulation and provides theoretical support for the clinical application of placental components, especially sub-cultured placental tissue-derived CM, in vascular tissue engineering and clinical treatments.
Collapse
Affiliation(s)
- Haiying Ma
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Shenglu Jiang
- Department of Pathophysiology, Zhangjiakou University, No.P19, Pingmen Street, Qiaoxi District, Zhangjiakou, 075000, Hebei Province, China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Jinfang Liu
- Department of Basic Medical Sciences, Basic Medical College, Shan Xi University of Traditional Chinese Medicine, No. 89, Section 1, Jinci Road, Taiyuan, 030024, Shanxi Province, China
| | - Xiaoyan Xu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Hua Zhu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Jun Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China. .,Shenyang Engineering Technology R&D Center of Cell Therapy CO.LTD, No. 400-8, Zhihui 2nd Street, Hunnan District, Shenyang, 110169, Liaoning Province, China.
| |
Collapse
|
22
|
Kalashnikova LA, Danilova MS, Gubanova MV, Dobrynina LA. [Cervical artery dissection in women: relationships with pregnancy and postpartum period]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:7-12. [PMID: 34874648 DOI: 10.17116/jnevro20211211017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study the frequency of CeAD that developed during pregnancy or in post partum period among all CeADs in women; to study the course of pregnancy in women with prior CeAD. MATERIAL AND METHODS 162 women (mean age 37.1±4.1 years) with CeAD we examined at the Research Center of Neurology (Moscow), 98% women were studied during last 15 years. 140 women were of childbearing age (≤45 years, mean age - 35±2.8 years). All patients were interviewed whether or not CeAD occurred during pregnancy or in post partum period (CeADPPP). Obstetric history before and after CeAD was studied in 57 women of childbearing age (average age - 35.9±7.3 years at CeAD development). RESULTS CeADPPP developed in 6 out of 162 all female patients (3.7%) or of 140 childbearing age patients (4.3%). It occurred 2-6 months (4 patients) and 10 days after delivery (1 patient), or on the 25th week of pregnancy (1 patient). CeADPPP patients were younger than patients with CeAD out of pregnancy or postpartum period (29.8±8 years vs 35.1±6.7 years, p>0.05). CeADPPP in comparison with CeAD outside these periods more often involved internal carotid artery (ICA) (50% vs 35%, p=0.666), more often occurred in 2-3 arteries (50% vs 31%, p=0.386) and more often was accompanied by dissecting aneurysm development (50% vs 8%, p=0.013). After CeAD, 18 out of 57 patients in whom obstetric history was studied, including 3 patients with postpartum dissection had 29 pregnancies. The pregnancy outcomes were as follows: childbirth (17 pregnancies, 59%), fetal loss (8 pregnancies, 27%) and medical abortion (4 pregnancies, 14%). Delivery occurred on average 4.5±2.061 years after CeAD in women aged 33.0±4.25 years (cesarean section - 15 patients). Fetal loss occurred at 7.4±3.5 weeks of pregnancy in women aged 37.6±3.13 years on average 2.7±1.4 years after CeAD. Fetal loss frequency after CeAD was higher than before it (27% vs 7%, p=0.016). There were no CeAD recurrences during pregnancy and postpartum period in women who had previously undergone CeAD. CONCLUSION CeADPPP frequency among all dissections in women is 3.7-4.3%. The risk of CeAD recurrence during pregnancy or the postpartum period after prior CeAD is very low. The risk of fetal loss during 2.7±1.4 years after CeAD is higher than before it (27% vs 7%). Hormonal and growth factors associated with pregnancy and the postpartum period is suggested to contribute to cervical artery wall damage. It is possible that the prolonged action of some of these factors may disrupt the placental vessels formation, predisposing to miscarriage.
Collapse
|
23
|
Raez-Villanueva S, Perono GA, Jamshed L, Thomas PJ, Holloway AC. Effects of dibenzothiophene, a sulfur-containing heterocyclic aromatic hydrocarbon, and its alkylated congener, 2,4,7-trimethyldibenzothiophene, on placental trophoblast cell function. J Appl Toxicol 2020; 41:1367-1379. [PMID: 33314207 DOI: 10.1002/jat.4128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/19/2020] [Accepted: 12/04/2020] [Indexed: 12/31/2022]
Abstract
Worldwide demand for petroleum products has resulted in increased oil and gas activities in many countries. Conventional and unconventional oil and gas extraction, production, and transport lead to increased levels of petroleum-derived polycyclic aromatic hydrocarbons (PAHs) in the environment. PAH exposure has profound effects on reproduction by affecting pathways involved in placental trophoblast cell function and impairing normal placental development and function-key contributors to reproductive success. However, other components found in petroleum and wastewaters from oil and gas extraction, including the sulfur-containing heterocyclic aromatic compounds such as dibenzothiophene (DBT) and its alkylated derivatives, may also impact reproductive success. The goal of this study was to examine the effect of exposure to DBT, a compound commonly detected in the environment, and one of its alkylated analogues, 2,4,7-trimethyldibenzothiophene (2,4,7-DBT), on steroidogenic and angiogenic pathways critical for mammalian development in placental trophoblast cells (HTR-8/SVneo cells). 2,4,7-DBT but not DBT increased estradiol output in association with increased tube-like formation (surrogate for angiogenesis). These changes in angiogenesis did not appear to be related to altered expression of the key placental angiogenic gene targets (ANGPTL4, VEGFA, and PGF). Neither compound showed a concentration related effect on progesterone synthesis or its receptor expression. Our results suggest that 2,4,7-DBT can disrupt key pathways important for placental trophoblast function and highlight the importance of determining the impact of exposure to both parent and alkylated compounds. Further, these data suggest that exposure to sulfur-containing heterocyclic aromatic compounds may lead to placental dysfunction and impact reproductive success at environmentally relevant levels.
Collapse
Affiliation(s)
| | - Genevieve A Perono
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Laiba Jamshed
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Philippe J Thomas
- Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa, Ontario, Canada
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Maternal Docosahexaenoic Acid Status during Pregnancy and Its Impact on Infant Neurodevelopment. Nutrients 2020; 12:nu12123615. [PMID: 33255561 PMCID: PMC7759779 DOI: 10.3390/nu12123615] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Dietary components are essential for the structural and functional development of the brain. Among these, docosahexaenoic acid, 22:6n-3 (DHA), is critically necessary for the structure and development of the growing fetal brain in utero. DHA is the major n-3 long-chain polyunsaturated fatty acid in brain gray matter representing about 15% of all fatty acids in the human frontal cortex. DHA affects neurogenesis, neurotransmitter, synaptic plasticity and transmission, and signal transduction in the brain. Data from human and animal studies suggest that adequate levels of DHA in neural membranes are required for maturation of cortical astrocyte, neurovascular coupling, and glucose uptake and metabolism. Besides, some metabolites of DHA protect from oxidative tissue injury and stress in the brain. A low DHA level in the brain results in behavioral changes and is associated with learning difficulties and dementia. In humans, the third trimester-placental supply of maternal DHA to the growing fetus is critically important as the growing brain obligatory requires DHA during this window period. Besides, DHA is also involved in the early placentation process, essential for placental development. This underscores the importance of maternal intake of DHA for the structural and functional development of the brain. This review describes DHA’s multiple roles during gestation, lactation, and the consequences of its lower intake during pregnancy and postnatally on the 2019 brain development and function.
Collapse
|
25
|
Jing G, Yao J, Dang Y, Liang W, Xie L, Chen J, Li Z. The role of β-HCG and VEGF-MEK/ERK signaling pathway in villi angiogenesis in patients with missed abortion. Placenta 2020; 103:16-23. [PMID: 33068962 DOI: 10.1016/j.placenta.2020.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To analyze the effects of the Human Chorionic Gonadotropin beta (β-hCG) and the VEGF-MEK/ERK signaling pathway on villi angiogenesis in early missed abortion. METHODS A total of 12 cases of women with missed abortion and 12 cases of women who had induced abortion voluntarily without any disease were included in the present study. The age, pregnancy time and gestation period in the control group corresponded to the missed abortion group. Wes Simple Western system and qRT-PCR were used to detect the expression of VEGF-MEK/ERK signaling pathway related proteins and genes in villous. Radioimmunoassay and Enzyme-linked immunosorbent assay were used to detect β-hCG and VEGF levels in serum. The microvascular density (MVD) in villous tissue was analyzed by immunohistochemical staining. RESULTS The levels of β-hCG and VEGF in serum, the expression of VEGF-MEK/ERK signaling pathway and MVD in villous tissue of the missed abortion group were lower than those of the control group. In addition, compared with the control group, the layers of trophoblasts of the villous tissue in the missed abortion group became thinner significantly, the number of cells reduced, the cell structures were disorganized, and parts of the trophoblast cells were absent. Correlational analysis showed that the protein expression of ERK1/2 was positively correlated with MVD in missed abortion group. CONCLUSIONS Our results reveal that decreased production of β-hCG in early pregnant women could down-regulate the expression of VEGF-MEK/ERK signal pathway, then reduce angiogenesis and eventually leading to the abnormal angiogenesis of villous, which may be an important mechanism of missed abortion.
Collapse
Affiliation(s)
- Guangzhuang Jing
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Jianling Yao
- Maternal and Child Health Hospital of Jingning Country, Lanzhou, 743400, China
| | - Yuhui Dang
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Weitao Liang
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Li'ao Xie
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Jia Chen
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Zhilan Li
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
26
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
27
|
Alfaidy N, Baron C, Antoine Y, Reynaud D, Traboulsi W, Gueniffey A, Lamotte A, Melloul E, Dunand C, Villaret L, Bessonnat J, Mauroy C, Boueihl T, Coutton C, Martinez G, Hamamah S, Hoffmann P, Hennebicq S, Brouillet S. Prokineticin 1 is a new biomarker of human oocyte competence: expression and hormonal regulation throughout late folliculogenesis. Biol Reprod 2020; 101:832-841. [PMID: 31276578 DOI: 10.1093/biolre/ioz114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/08/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Nadia Alfaidy
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
| | - Chloé Baron
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
- INSERM U1203, Equipe "Développement Embryonnaire Précoce Humain et Pluripotence", Institut de Médecine Régénératrice et de Biothérapie, Hôpital Saint-Eloi, Montpellier 34295, France
| | - Yannick Antoine
- INSERM U1203, Equipe "Développement Embryonnaire Précoce Humain et Pluripotence", Institut de Médecine Régénératrice et de Biothérapie, Hôpital Saint-Eloi, Montpellier 34295, France
| | - Déborah Reynaud
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
| | - Wael Traboulsi
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
| | - Aurore Gueniffey
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Anna Lamotte
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Eve Melloul
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Camille Dunand
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Laure Villaret
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Julien Bessonnat
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Charlotte Mauroy
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Thomas Boueihl
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Charles Coutton
- Université Grenoble-Alpes, Inserm, Institute for Advanced Biosciences (IAB), équipe Génétique Epigénétique et Thérapie de l'Infertilité (GETI), 38000, Grenoble, France
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple Enfant, Département de Génétique et Procréation, Laboratoire de Génétique Chromosomique, 38700, La Tronche, France
| | - Guillaume Martinez
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple Enfant, Département de Génétique et Procréation, Laboratoire de Génétique Chromosomique, 38700, La Tronche, France
| | - Samir Hamamah
- INSERM U1203, Equipe "Développement Embryonnaire Précoce Humain et Pluripotence", Institut de Médecine Régénératrice et de Biothérapie, Hôpital Saint-Eloi, Montpellier 34295, France
- CHU Montpellier, ART/PGD Division, Hôpital Arnaud de Villeneuve, Montpellier 34295, France
| | - Pascale Hoffmann
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
| | - Sylviane Hennebicq
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
- Université Grenoble-Alpes, Inserm, Institute for Advanced Biosciences (IAB), équipe Génétique Epigénétique et Thérapie de l'Infertilité (GETI), 38000, Grenoble, France
| | - Sophie Brouillet
- Université Grenoble-Alpes, Inserm, Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut de Biosciences et Biotechnologies de Grenoble (BIG), Laboratoire Biologie du Cancer et de l'Infection (BCI), 38000, Grenoble, France
- Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation- Centre d'étude et de conservation des œufs et du sperme humains (CECOS), 38700, La Tronche, France
- INSERM U1203, Equipe "Développement Embryonnaire Précoce Humain et Pluripotence", Institut de Médecine Régénératrice et de Biothérapie, Hôpital Saint-Eloi, Montpellier 34295, France
| |
Collapse
|
28
|
Goryszewska E, Kaczynski P, Balboni G, Waclawik A. Prokineticin 1-prokineticin receptor 1 signaling promotes angiogenesis in the porcine endometrium during pregnancy†. Biol Reprod 2020; 103:654-668. [PMID: 32355954 DOI: 10.1093/biolre/ioaa066] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/20/2020] [Accepted: 04/29/2020] [Indexed: 01/16/2023] Open
Abstract
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-17β, 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.
Collapse
Affiliation(s)
- Ewelina Goryszewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Piotr Kaczynski
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Gianfranco Balboni
- Unit of Pharmaceutical, Pharmacological and Nutraceutical Sciences, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
29
|
Ujvari D, Graells Brugalla C, Hirschberg AL. Dihydrotestosterone potentiates insulin to up-regulate prokineticin-1 in decidualizing human endometrial stromal cells. J Cell Mol Med 2020; 24:3242-3245. [PMID: 31991505 PMCID: PMC7077604 DOI: 10.1111/jcmm.14923] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 11/29/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | | | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
Umapathy A, Chamley LW, James JL. Reconciling the distinct roles of angiogenic/anti-angiogenic factors in the placenta and maternal circulation of normal and pathological pregnancies. Angiogenesis 2019; 23:105-117. [DOI: 10.1007/s10456-019-09694-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/26/2019] [Indexed: 01/03/2023]
|
31
|
Kelley AS, Puttabyatappa M, Ciarelli JN, Zeng L, Smith YR, Lieberman R, Pennathur S, Padmanabhan V. Prenatal Testosterone Excess Disrupts Placental Function in a Sheep Model of Polycystic Ovary Syndrome. Endocrinology 2019; 160:2663-2672. [PMID: 31436841 PMCID: PMC6804485 DOI: 10.1210/en.2019-00386] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common condition of reproductive-aged women. In a well-validated sheep model of PCOS, testosterone (T) treatment of pregnant ewes culminated in placental insufficiency and intrauterine growth restriction of offspring. The purpose of this study was to explore specific mechanisms by which T excess compromises placental function in early, mid, and late gestation. Pregnant Suffolk sheep received T propionate 100 mg intramuscularly or control vehicle twice weekly from gestational days (GD) 30 to 90 (term = 147 days). Placental harvest occurred at GD 65, 90, and 140. Real-time RT-PCR was used to assess transcript levels of proinflammatory (TNF, IL1B, IL6, IL8, monocyte chemoattractant protein-1/chemokine ligand 2, cluster of differentiation 68), antioxidant (glutathione reductase and superoxide dismutase 1 and 2), and angiogenic [vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1α (HIF1A)] genes. Lipid accumulation was assessed using triglyceride assays and Oil Red O staining. Placental measures of oxidative and nitrative stress included the thiobarbituric acid reactive substance assay and high-pressure liquid chromatography. Tissue fibrosis was assessed with Picrosirius Red staining. Student t tests and Cohen effect-size analyses were used for statistical analysis. At GD 65, T-treated placentomes showed increased lipid accumulation and collagen deposition. Notable findings at GD 90 were a significant increase in HIF1A expression and a large effect increase in VEGF expression. At GD 140, T-treated placentomes displayed large effect increases in expression of hypoxia and inflammatory markers. In summary, T treatment during early pregnancy induces distinct gestational age-specific effects on the placental milieu, which may underlie the previously observed phenotype of placental insufficiency.
Collapse
Affiliation(s)
- Angela S Kelley
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | | | - Joseph N Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Lixia Zeng
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yolanda R Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Richard Lieberman
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Correspondence: Vasantha Padmanabhan, PhD, Department of Pediatrics, University of Michigan, 7510 MSRB 1, 1500 West Medical Center Drive, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
32
|
Meng L, Yang H, Jin C, Quan S. miR‑28‑5p suppresses cell proliferation and weakens the progression of polycystic ovary syndrome by targeting prokineticin‑1. Mol Med Rep 2019; 20:2468-2475. [PMID: 31322191 DOI: 10.3892/mmr.2019.10446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/30/2019] [Indexed: 11/05/2022] Open
Abstract
Prokineticin‑1 (PROK1) serves important roles in the pathogenesis of polycystic ovary syndrome (PCOS); however, the association between microRNA (miR)‑28‑5p and PROK1 remains unclear. In the present study, the roles of miR‑28‑5p and PROK1, and their interaction in PCOS were investigated. Rat ovary granule cells were transfected with miR‑28‑5p mimics, and PROK1 expression levels were measured by reverse transcription‑quantitative PCR and western blotting. A dual‑luciferase reporter assay was performed to determine the association between miR‑28‑5p and PROK1. Additionally, pcDNA‑PROK1 was co‑transfected into rat ovary granule cells with miR‑28‑5p mimics. Cell proliferation, apoptosis, cell cycle and the expression of signaling proteins were investigated using Cell Counting Kit‑8 assays, 5‑ethynyl‑2'‑deoxyuridine staining, flow cytometry and western blotting, respectively. PROK1 expression was suppressed in rat ovary granule cells by miR‑28‑5p mimics, but upregulated following transfection with miR‑28‑5p inhibitors. The dual‑luciferase reporter assay revealed that miR‑28‑5p binds to the 3'‑untranslated region of PROK1. Proliferation activity was increased in PROK1‑overexpressing cells; this effect was eliminated by co‑transfection with miR‑28‑5p mimics. PROK1‑overexpressing rat ovary granule cells exhibited significantly suppressed cell apoptosis and a decreased number of cells in G1; miR‑28‑5p mimics reversed these effects. Western blotting revealed that the PI3K/AKT/mTOR signaling pathway was activated by PROK1. The present results suggested that miR‑28‑5p attenuated the progression of PCOS by targeting PROK1, which may promote the pathogenesis of PCOS via the PI3K/AKT/mTOR pathway, indicating that the miR‑28‑5p/PROK1 axis may be a potential therapeutic target for patients with PCOS.
Collapse
Affiliation(s)
- Lyuhe Meng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Congcong Jin
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Song Quan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
33
|
Wang CY, Tsai PY, Chen TY, Tsai HL, Kuo PL, Su MT. Elevated miR-200a and miR-141 inhibit endocrine gland-derived vascular endothelial growth factor expression and ciliogenesis in preeclampsia. J Physiol 2019; 597:3069-3083. [PMID: 31026335 DOI: 10.1113/jp277704] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/09/2019] [Indexed: 12/23/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yu Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ling Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
34
|
Benfateh M, Cissoko S, Boufettal H, Feige JJ, Samouh N, Aboussaouira T, Benharouga M, Alfaidy N. Risk factors and poor prognostic factors of preeclampsia in Ibn Rochd University Hospital of Casablanca: about 401 preeclamptic cases. Pan Afr Med J 2018; 31:225. [PMID: 31447983 PMCID: PMC6691300 DOI: 10.11604/pamj.2018.31.225.14401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/14/2018] [Indexed: 11/11/2022] Open
Abstract
Preeclampsia is a gestational pathology that complicates 2 to 8% of pregnancies and is one of the major causes of maternal and fetal morbidity and mortality worldwide. The aim of this work was to study the epidemiological profile of preeclampsia in Casablanca and to identify risk factors as well as factors of poor maternal and fetal prognosis. 401 preeclamptic cases were collected in the gynecology-obstetrics "C" Service of Lalla Meryem Maternity of Ibn Rochd University Hospital of Casablanca (2010-2011) were included in this study and a statistical analysis with the SPSS software version (16.0) was performed. We used the Chi-2 test to analyze qualitative variables and Student's test and ANOVA (analysis of variance) for quantitative variables. The incidence of preeclampsia was (7.1%). The epidemiological profile was that of a primipara (57.6%), average age 30 years and (66.8%) of pregnancies were not followed. Multiparity was a factor of poor maternal prognosis (p = 0.007). The low gestational age and no prenatal care were factors of maternal as well as fetal prognosis. Risk factors frequently found in our patients were obesity (15.21%) and chronic hypertension (5.73%) as vascular-renal history; abortion (16.46%) and perinatal death (5.24%) as obstetric history. Preeclampsia is a common obstetric pathology in our context. Better prenatal care and early diagnosis could reduce its incidence.
Collapse
Affiliation(s)
- Meriem Benfateh
- Unité de Culture Cellulaire, CED, Faculté de Médecine et de Pharmacie, Hassan II University of Casablanca, Maroc
| | - Souadou Cissoko
- Unité de Culture Cellulaire, CED, Faculté de Médecine et de Pharmacie, Hassan II University of Casablanca, Maroc
| | - Houssine Boufettal
- Service C de Gynécologie, CHU Ibn Rochd, Faculté de Médecine et de Pharmacie, Hassan II University of Casablanca, Maroc
| | - Jean-Jacques Feige
- Institut de Biosciences et Biotechnologie du CEA de Grenoble, Unité U1036 INSERM, Grenoble, France
| | - Naima Samouh
- Service C de Gynécologie, CHU Ibn Rochd, Faculté de Médecine et de Pharmacie, Hassan II University of Casablanca, Maroc
| | - Touria Aboussaouira
- Unité de Culture Cellulaire, CED, Faculté de Médecine et de Pharmacie, Hassan II University of Casablanca, Maroc
| | - Mohamed Benharouga
- Commissariat à l'Energie Atomique (CEA), DSV-iRTSV, 17 rue des Martyrs, France-Université Grenoble-Alpes, Centre National de la Recherche Scientifique, UMR 5249, 38054 Grenoble Cedex 9, France
| | - Nadia Alfaidy
- Institut de Biosciences et Biotechnologie du CEA de Grenoble, Unité U1036 INSERM, Grenoble, France
| |
Collapse
|
35
|
Negri L, Ferrara N. The Prokineticins: Neuromodulators and Mediators of Inflammation and Myeloid Cell-Dependent Angiogenesis. Physiol Rev 2018. [PMID: 29537336 DOI: 10.1152/physrev.00012.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mammalian prokineticins family comprises two conserved proteins, EG-VEGF/PROK1 and Bv8/PROK2, and their two highly related G protein-coupled receptors, PKR1 and PKR2. This signaling system has been linked to several important biological functions, including gastrointestinal tract motility, regulation of circadian rhythms, neurogenesis, angiogenesis and cancer progression, hematopoiesis, and nociception. Mutations in PKR2 or Bv8/PROK2 have been associated with Kallmann syndrome, a developmental disorder characterized by defective olfactory bulb neurogenesis, impaired development of gonadotropin-releasing hormone neurons, and infertility. Also, Bv8/PROK2 is strongly upregulated in neutrophils and other inflammatory cells in response to granulocyte-colony stimulating factor or other myeloid growth factors and functions as a pronociceptive mediator in inflamed tissues as well as a regulator of myeloid cell-dependent tumor angiogenesis. Bv8/PROK2 has been also implicated in neuropathic pain. Anti-Bv8/PROK2 antibodies or small molecule PKR inhibitors ameliorate pain arising from tissue injury and inhibit angiogenesis and inflammation associated with tumors or some autoimmune disorders.
Collapse
Affiliation(s)
- Lucia Negri
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| | - Napoleone Ferrara
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| |
Collapse
|
36
|
Erzincan SG, Varol FG, Inan C, Sayin NC. Relationship between second-trimester amniotic fluid levels of Prokineticin-1 and Matrix Metalloproteinase-2 with adverse pregnancy outcome. Placenta 2018; 62:25-27. [PMID: 29405963 DOI: 10.1016/j.placenta.2017.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/02/2017] [Accepted: 12/17/2017] [Indexed: 11/17/2022]
Abstract
To investigate the levels of Prokineticin-1 (PROK1) and matrix metalloproteinase-2 (MMP-2) in second-trimester amniotic fluid (AF). AF samples were investigated in 81 patients. AF-PROK1 and AF-MMP-2 were not significantly associated with adverse pregnancy outcomes (preeclampsia, intrauterine growth retardation, spontaneous preterm birth, gestational diabetes, gestational hypertension). AF-PROK1 levels in patients with abnormal first-trimester screening were significantly higher than those who underwent amniocentesis due to abnormal second-trimester screening tests (p = .04). AF-PROK1 or AF-MMP-2 do not have a role in the prediction of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Selen G Erzincan
- Trakya University, Faculty of Medicine, Department of Obstetrics & Gynecology, Division of Perinatology, Edirne, Turkey.
| | - Fusun G Varol
- Trakya University, Faculty of Medicine, Department of Obstetrics & Gynecology, Division of Perinatology, Edirne, Turkey.
| | - Cihan Inan
- Trakya University, Faculty of Medicine, Department of Obstetrics & Gynecology, Division of Perinatology, Edirne, Turkey.
| | - N Cenk Sayin
- Trakya University, Faculty of Medicine, Department of Obstetrics & Gynecology, Division of Perinatology, Edirne, Turkey.
| |
Collapse
|
37
|
Chang X, Li H, Li Y, He Q, Yao J, Duan T, Wang K. RhoA/MLC signaling pathway is involved in Δ⁹-tetrahydrocannabinol-impaired placental angiogenesis. Toxicol Lett 2018; 285:148-155. [PMID: 29307655 DOI: 10.1016/j.toxlet.2017.12.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/28/2017] [Accepted: 12/31/2017] [Indexed: 01/26/2023]
Abstract
Cannabis is a widely used illicit drug and its abuse during pregnancy has been related to adverse reproductive outcomes. In addition, placental angiogenesis is considered to be responsible for the transport of nutrients critical for placental development and fetal growth. The purpose of this study is to determine the effects of Δ⁹-tetrahydrocannabinol (THC), the major component of cannabis, on placental angiogenesis, involving endothelial cell (EC) proliferation, migration and tube formation. Here, we observe dramatic alterations in placental vascular network of cannabis users correlated with an impaired HUVE cell proliferation, migration and tube formation after treated with THC. Mechanistically, the activity of RhoA/MLC is involved in the THC-impaired EC migration and angiogenesis. To further analyze the role of cannabis in mice placental and embryonic development, we inject pregnant mice with THC daily. This treatment results in an altered placental microvasculature, accompanied by the decreased expression of CD31 and activity of RhoA/MLC. Taken together, these findings identify THC plays a pivotal role in impairing placental angiogenesis potentially via RhoA/MLC signaling nexus.
Collapse
Affiliation(s)
- Xinwen Chang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China
| | - Hua Li
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China
| | - Yuhong Li
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China
| | - Julei Yao
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China
| | - Tao Duan
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China; Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China.
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, PR China.
| |
Collapse
|
38
|
Korzeniewski SJ, Allred EN, Joseph RM, Heeren T, Kuban KC, O’Shea TM, Leviton A. Neurodevelopment at Age 10 Years of Children Born <28 Weeks With Fetal Growth Restriction. Pediatrics 2017; 140:peds.2017-0697. [PMID: 29030525 PMCID: PMC5654396 DOI: 10.1542/peds.2017-0697] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES We sought to evaluate the relationships between fetal growth restriction (FGR) (both severe and less severe) and assessments of cognitive, academic, and adaptive behavior brain function at age 10 years. METHODS At age 10 years, the Extremely Low Gestational Age Newborns Cohort Study assessed the cognitive function, academic achievement, social-communicative function, psychiatric symptoms, and overall quality of life of 889 children born before 28 weeks' gestation. A pediatric epileptologist also interviewed parents as part of a seizure evaluation. The 52 children whose birth weight z scores were <-2 were classified as having severe FGR, and the 113 whose birth weight z scores were between -2 and -1 were considered to have less severe FGR. RESULTS The more severe the growth restriction in utero, the lower the level of function on multiple cognitive and academic achievement assessments performed at age 10 years. Growth-restricted children were also more likely than their extremely preterm peers to have social awareness impairments, autistic mannerisms, autism spectrum diagnoses, difficulty with semantics and speech coherence, and diminished social and psychosocial functioning. They also more frequently had phobias, obsessions, and compulsions (according to teacher, but not parent, report). CONCLUSIONS Among children born extremely preterm, those with severe FGR appear to be at increased risk of multiple cognitive and behavioral dysfunctions at age 10 years, raising the possibility that whatever adversely affected their intrauterine growth also adversely affected multiple domains of cognitive and neurobehavioral development.
Collapse
Affiliation(s)
- Steven J. Korzeniewski
- Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, Michigan;,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan
| | - Elizabeth N. Allred
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts;,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts
| | | | - Tim Heeren
- Department of Biostatistics, School of Public Health
| | - Karl C.K. Kuban
- Boston University, Boston, Massachusetts;,Departments of Pediatrics, Boston Medical Center, Boston, Massachusetts; and
| | - T. Michael O’Shea
- Department of Pediatrics, Wake Forest University, Winston-Salem, North Carolina
| | - Alan Leviton
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts;,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts
| | | |
Collapse
|
39
|
Olivera-Severo D, Uberti AF, Marques MS, Pinto MT, Gomez-Lazaro M, Figueiredo C, Leite M, Carlini CR. A New Role for Helicobacter pylori Urease: Contributions to Angiogenesis. Front Microbiol 2017; 8:1883. [PMID: 29021786 PMCID: PMC5623709 DOI: 10.3389/fmicb.2017.01883] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/14/2017] [Indexed: 12/29/2022] Open
Abstract
Helicobacter pylori is a pathogen involved in gastric diseases such as ulcers and carcinomas. H. pylori's urease is an important virulence factor produced in large amounts by this bacterium. In previous studies, we have shown that this protein is able to activate several cell types like neutrophils, monocytes, platelets, endothelial cells, and gastric epithelial cells. Angiogenesis is a physiological process implicated in growth, invasion and metastization of tumors. Here, we have analyzed the angiogenic potential of H. pylori urease (HPU) in gastric epithelial cells. No cytotoxicity was observed in AGS, Kato-III, and MKN28 gastric cell lines treated with 300 nM HPU, as evaluated by the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. As we previously reported in neutrophils, treatment with 300 nM HPU also had an anti-apoptotic effect in gastric epithelial cells leading to a 2.2-fold increase in the levels of Bcl-XL after 6 h, and a decrease of 80% in the content of BAD, after 48 h, two mitochondrial proteins involved in regulation of apoptosis. Within 10 min of exposure, HPU is rapidly internalized by gastric epithelial cells. Treatment of the gastric cells with methyl-β-cyclodextrin abolished HPU internalization suggesting a cholesterol-dependent process. HPU induces the expression of pro-angiogenic factors and the decrease of expression of anti-angiogenic factors by AGS cells. The angiogenic activity of HPU was analyzed using in vitro and in vivo models. HPU induced formation of tube-like structures by human umbilical vascular endothelial cells in a 9 h experiment. In the chicken embryo chorioallantoic membrane model, HPU induced intense neo-vascularization after 3 days. In conclusion, our results indicate that besides allowing bacterial colonization of the gastric mucosa, H. pylori's urease triggers processes that initiate pro-angiogenic responses in different cellular models. Thus, this bacterial urease, a major virulence factor, may also play a role in gastric carcinoma development.
Collapse
Affiliation(s)
- Deiber Olivera-Severo
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil.,Biology Department, Universidade Regional Integrada do Alto Uruguai e das Missões, São Luiz Gonzaga, Brazil
| | - Augusto F Uberti
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil.,Institute of Biology, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Miguel S Marques
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Marta T Pinto
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Maria Gomez-Lazaro
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - Céu Figueiredo
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Marina Leite
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Célia R Carlini
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil.,Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
40
|
Ujvari D, Jakson I, Oldmark C, Attarha S, Alkasalias T, Salamon D, Gidlöf S, Hirschberg AL. Prokineticin 1 is up-regulated by insulin in decidualizing human endometrial stromal cells. J Cell Mol Med 2017; 22:163-172. [PMID: 28782224 PMCID: PMC5742737 DOI: 10.1111/jcmm.13305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/03/2017] [Indexed: 12/21/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ivika Jakson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Oldmark
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sanaz Attarha
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Twana Alkasalias
- Department of Microbiology, Tumour, and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biology, College of Science, Salahaddin University, Irbil, Kurdistan-Iraq
| | - Daniel Salamon
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Gidlöf
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
41
|
Inan C, Varol FG, Erzincan SG, Uzun I, Sutcu H, Sayin NC. Use of prokineticin-1 (PROK1), pregnancy-associated plasma protein A (PAPP-A) and PROK1/PAPP-A ratio to predict adverse pregnancy outcomes in the first trimester: a prospective study. J Matern Fetal Neonatal Med 2017; 31:2685-2692. [PMID: 28675948 DOI: 10.1080/14767058.2017.1351536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION To compare the predictive effectiveness levels of prokineticin-1 (PROK1), pregnancy-associated plasma protein A (PAPP-A) and the PROK1/PAPP-A ratio in the first trimester for preeclampsia (PE), foetal growth restriction (FGR), gestational diabetes mellitus (GDM) and spontaneous preterm birth (SPB). MATERIALS AND METHODS A total of randomly selected 162 pregnant women were included. Peripheral blood samples were obtained between 110/7 and 136/7 gestational weeks (GWs). All women were followed throughout the pregnancy and classified into five groups as having PE, FGR, GDM, SPB and uncomplicated pregnancies. The cut-off levels of the markers were identified to predict adverse outcomes. RESULTS PROK1 predicted PE with 83.3% sensitivity, 85.7% specificity at a value of >293.4 pg/mL; at a value of >260.2 pg/mL, PROK1 predicted FGR with 85.7% sensitivity, 72.5% specificity in the first trimester. The area under receiver operating characteristic (ROC) curve of PAPP-A was lower than that of PROK1 and PROK1/PAPP-A in differentiating PE and FGR from the uncomplicated group (p < .001). PROK1 levels and the PROK1/PAPP-A ratios in the SPB and GDM groups were lower than in the uncomplicated group (p < .01). CONCLUSIONS Elevated PROK1 in the first trimester is a more effective marker than PAPP-A in the prediction of PE and FGR. Lower PROK1 levels are associated with the development of SPB and GDM.
Collapse
Affiliation(s)
- Cihan Inan
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| | - Fusun Gulizar Varol
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| | - Selen Gursoy Erzincan
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| | - Isil Uzun
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| | - Havva Sutcu
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| | - N Cenk Sayin
- a Trakya University Faculty of Medicine, Obstetrics and Gynecology , Edirne , Turkey
| |
Collapse
|
42
|
|
43
|
Stangret A, Skoda M, Wnuk A, Pyzlak M, Szukiewicz D. Mild anemia during pregnancy upregulates placental vascularity development. Med Hypotheses 2017; 102:37-40. [PMID: 28478827 DOI: 10.1016/j.mehy.2017.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/05/2017] [Indexed: 11/16/2022]
Abstract
The connection between maternal hematological status and pregnancy outcome has been shown by many independent researchers. Attention was initially focused on the adverse effects of moderate and severe anemia. Interestingly, some studies revealed that mild anemia was associated with optimal fetal development and was not affecting pregnancy outcome. The explanation for this phenomenon became a target for scientists. Hemodilution, physiologic anemia and relative decrease in hemoglobin concentration are the changes observed during pregnancy but they do not explain the reasons for the positive influence of mild anemia on a fetomaternal unit. It is hypothesized that hemodilution facilitates placental perfusion because blood viscosity is reduced. Subsequently, it may lead to a decline in hemoglobin concentration. Anemia from its definition implies decreased oxygen carrying capacity of the blood and can result in hypoxemia and even hypoxia, which is a common factor inducing new blood vessels formation. Therefore, we raised the hypothesis that the lowered hemoglobin concentration during pregnancy may upregulate vascular growth factor receptors expression such as VEGFR-1 (Flt-1) and VEGFR-2 (FLK-1/KDR). Consecutively, increased fetoplacental vasculogenesis and angiogenesis provide further expansion of vascular network development, better placental perfusion and hence neither fetus nor the mother are affected.
Collapse
Affiliation(s)
- A Stangret
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland.
| | - M Skoda
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - A Wnuk
- Chair and Department of Obstetrics, Gynecology, and Oncology, 2nd Faculty of Medical University of Warsaw, Poland
| | - M Pyzlak
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| | - D Szukiewicz
- Chair and Department of General and Experimental Pathology with Centre for Preclinical Research and Technology, Medical University of Warsaw, Poland
| |
Collapse
|
44
|
Su MT, Tsai PY, Tsai HL, Chen YC, Kuo PL. miR-346 and miR-582-3p-regulated EG-VEGF expression and trophoblast invasion via matrix metalloproteinases 2 and 9. Biofactors 2017; 43:210-219. [PMID: 27619846 DOI: 10.1002/biof.1325] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/14/2016] [Accepted: 08/09/2016] [Indexed: 12/31/2022]
Abstract
Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an important regulator for embryo implantation and placental development, and is clinically associated with several obstetric disorders related to insufficient or inappropriate trophoblast invasion, such as recurrent abortion, preeclampsia, and intrauterine fetal growth restriction. This study was performed to identify the microRNAs targeting EG-VEGF, and evaluate the regulatory effect on trophoblast biology. miR-346 and miR-582-3p were initially identified via bioinformatic tools, and their specific binding sites on the EG-VEGF 3'UTR were further confirmed using dual luciferase and a co-transfection assays. miR-346 and miR-582-3p were demonstrated not only to suppress EG-VEGF expression, but also inhibit trophoblast invasion and migration in the JAR and HTR-8/SVneo cell lines. We further evaluated the effect of microRNAs in HTR-8/SVneo cells coexpressing EG-VEGF and miR-346 or miR-582-3p on matrix metalloproteinase (MMP 2 and MMP 9) and the tissue inhibitors of metalloproteinase (TIMP 1 and TIMP 2) using RT-PCR, western blotting and gelatin zymography. TIMP 1 and TIMP 2 were not affected by the two microRNAs, whereas the expressions and activities of MMP 2 and MMP 9 were significantly downregulated, which in turn inhibited the invasion ability of trophoblasts. In conclusion, miR-346 and miR-582-3p regulate EG-VEGF-induced trophoblast invasion through repressing MMP 2 and MMP 9, and may become novel diagnostic biomarkers or therapeutic targets for EG-VEGF-related obstetric disorders. © 2016 BioFactors, 43(2):210-219, 2017.
Collapse
Affiliation(s)
- Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ling Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chi Chen
- Department of Economics, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
45
|
Altmäe S, Segura MT, Esteban FJ, Bartel S, Brandi P, Irmler M, Beckers J, Demmelmair H, López-Sabater C, Koletzko B, Krauss-Etschmann S, Campoy C. Maternal Pre-Pregnancy Obesity Is Associated with Altered Placental Transcriptome. PLoS One 2017; 12:e0169223. [PMID: 28125591 PMCID: PMC5268451 DOI: 10.1371/journal.pone.0169223] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/13/2016] [Indexed: 12/17/2022] Open
Abstract
Maternal obesity has a major impact on pregnancy outcomes. There is growing evidence that maternal obesity has a negative influence on placental development and function, thereby adversely influencing offspring programming and health outcomes. However, the molecular mechanisms underlying these processes are poorly understood. We analysed ten term placenta’s whole transcriptomes in obese (n = 5) and normal weight women (n = 5), using the Affymetrix microarray platform. Analyses of expression data were carried out using non-parametric methods. Hierarchical clustering and principal component analysis showed a clear distinction in placental transcriptome between obese and normal weight women. We identified 72 differentially regulated genes, with most being down-regulated in obesity (n = 61). Functional analyses of the targets using DAVID and IPA confirm the dysregulation of previously identified processes and pathways in the placenta from obese women, including inflammation and immune responses, lipid metabolism, cancer pathways, and angiogenesis. In addition, we detected new molecular aspects of obesity-derived effects on the placenta, involving the glucocorticoid receptor signalling pathway and dysregulation of several genes including CCL2, FSTL3, IGFBP1, MMP12, PRG2, PRL, QSOX1, SERPINE2 and TAC3. Our global gene expression profiling approach demonstrates that maternal obesity creates a unique in utero environment that impairs the placental transcriptome.
Collapse
Affiliation(s)
- Signe Altmäe
- Department of Women’s and Children’s Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
- Centre of Excellence for Paediatric Research EURISTIKOS and Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- * E-mail: (SA); (CC)
| | - Maria Teresa Segura
- Centre of Excellence for Paediatric Research EURISTIKOS and Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
| | | | - Sabine Bartel
- Division of Experimental Asthma Research, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Pilar Brandi
- Centre of Excellence for Paediatric Research EURISTIKOS and Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, Freising, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hans Demmelmair
- Ludwig-Maximilians-University of Munich, Dr. Hauner Children’s Hospital, University of Munich Medical Centre, Munich, Germany
| | - Carmen López-Sabater
- Department of Nutrition and Bromatology, School of Pharmacy, University of Barcelona, Spain
| | - Berthold Koletzko
- Ludwig-Maximilians-University of Munich, Dr. Hauner Children’s Hospital, University of Munich Medical Centre, Munich, Germany
| | - Susanne Krauss-Etschmann
- Division of Experimental Asthma Research, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Member of the German Center for Lung Research (DZL), Borstel, Germany
- Institute for Experimental Medicine, Christian-Albrechts-Universitaet zu Kiel, Kiel, Germany
- Comprehensive Pneumology Center, Ludwig Maximilians University Hospital and Helmholtz Zentrum München, Großhadern, Germany
| | - Cristina Campoy
- Centre of Excellence for Paediatric Research EURISTIKOS and Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Biohealth Institute of Granada, Granada, Spain
- * E-mail: (SA); (CC)
| |
Collapse
|
46
|
Corlan AS, Cîmpean AM, Jitariu AA, Melnic E, Raica M. Endocrine Gland-Derived Vascular Endothelial Growth Factor/Prokineticin-1 in Cancer Development and Tumor Angiogenesis. Int J Endocrinol 2017; 2017:3232905. [PMID: 28386275 PMCID: PMC5366234 DOI: 10.1155/2017/3232905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 12/26/2022] Open
Abstract
A lot of data suggests endocrine gland-derived vascular endothelial growth factor (EG-VEGF) to be restricted to endocrine glands and to some endocrine-dependent organs. Many evidences show that EG-VEGF stimulates angiogenesis and cell proliferation, although it is not a member of the VEGF family. At the time, a lot of data regarding the role of this growth factor in normal development are available. However, controversial results have been published in the case of pathological conditions and particularly in malignant tumors. Thus, our present paper has been focused on the role of EG-VEGF in normal tissues and various malignant tumors and their angiogenic processes.
Collapse
Affiliation(s)
- Ana Silvia Corlan
- Department of Endocrinology, “Vasile Goldis” University of Arad, Arad, Romania
| | - Anca Maria Cîmpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
- *Anca Maria Cîmpean:
| | - Adriana-Andreea Jitariu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
| | - Eugen Melnic
- Department of Pathology, “Nicolae Testemitanu” University of Medicine and Pharmacy, Chișinău, Moldova
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara, Romania
| |
Collapse
|
47
|
Prokineticin1 and pregnancy. ANNALES D'ENDOCRINOLOGIE 2016; 77:101-4. [PMID: 27172869 DOI: 10.1016/j.ando.2016.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 04/11/2016] [Indexed: 11/22/2022]
Abstract
Prokineticin 1 (PROK1), also called EG-VEGF, is a peptide of 86 amino acids with multiple biological functions. PROK1 acts via two G-protein coupled receptors: PROKR1 PROKR2. PROK1 is highly expressed in the placenta. This article reports the expression and the role of PROK1 during normal and pathological pregnancies: (i) during early pregnancy, PROK1 exhibits a peak of placental expression shortly before the establishment of the feto-maternal circulation; (ii) its receptors, PROKR1 PROKR2 are highly expressed in human placenta; (iii) its expression is increased by hypoxia; (iv) PROK1 inhibits extravillous trophoblasts migration and invasion and increases their proliferation and survival; (v) PROK1 is also a pro-angiogenic placental factor that increases microvascular placental endothelial cells proliferation, migration, invasion, and permeability. Circulating PROK1 levels are five times higher in pregnant women during the first trimester compared to the second and third trimesters. Also, its serum levels are higher in patients with preeclampsia (PE) and in patients with isolated intra-uterine growth restriction (IUGR). In mice, maintaining high level of PROK1 beyond its normal period of production (>10.5dpc) reproduces symptoms of PE. To date, our results demonstrated that PROK1 is a central factor of human placentation with direct roles both in the control of trophoblast invasion and villous growth. Thus, a failure in the expression of PROK1 and/or its receptor during pregnancy may contribute to the development of PE and/or IUGR. Besides theses original findings, we also report a direct role of this factor in parturition.
Collapse
|
48
|
Duttaroy AK. Docosahexaenoic acid supports feto-placental growth and protects cardiovascular and cognitive function: A mini review. EUR J LIPID SCI TECH 2016. [DOI: 10.1002/ejlt.201500496] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Asim K. Duttaroy
- Faculty of Medicine, Department of Nutrition; Institute of Basic Medical Sciences; University of Oslo; Norway
| |
Collapse
|
49
|
A Common Variant of PROK1 (V67I) Acts as a Genetic Modifier in Early Human Pregnancy through Down-Regulation of Gene Expression. Int J Mol Sci 2016; 17:ijms17020162. [PMID: 26828479 PMCID: PMC4783896 DOI: 10.3390/ijms17020162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/16/2016] [Accepted: 01/16/2016] [Indexed: 11/30/2022] Open
Abstract
PROK1-V67I has been shown to play a role as a modifier gene in the PROK1-PROKR system of human early pregnancy. To explore the related modifier mechanism of PROK1-V67I, we carried out a comparison study at the gene expression level and the cell function alternation of V67I, and its wild-type (WT), in transiently-transfected cells. We, respectively, performed quantitative RT-PCR and ELISA assays to evaluate the protein and/or transcript level of V67I and WT in HTR-8/SV neo, JAR, Ishikawa, and HEK293 cells. Transiently V67I- or WT-transfected HTR-8/SV neo and HEK293 cells were used to investigate cell function alternations. The transcript and protein expressions were down-regulated in all cell lines, ranging from 20% to 70%, compared with WT. There were no significant differences in the ligand activities of V67I and WT with regard to cell proliferation, cell invasion, calcium influx, and tubal formation. Both PROK1 alleles promoted cell invasion and intracellular calcium mobilization, whereas they had no significant effects on cell proliferation and tubal formation. In conclusion, the biological effects of PROK1-V67I on cell functions are similar to those of WT, and the common variant of V67I may act as a modifier in the PROK1-PROKR system through down-regulation of PROK1 expression. This study may provide a general mechanism that the common variant of V67I, modifying the disease severity of PROK1-related pathophysiologies.
Collapse
|
50
|
Ferulic Acid Exerts Anti-Angiogenic and Anti-Tumor Activity by Targeting Fibroblast Growth Factor Receptor 1-Mediated Angiogenesis. Int J Mol Sci 2015; 16:24011-31. [PMID: 26473837 PMCID: PMC4632735 DOI: 10.3390/ijms161024011] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/21/2015] [Accepted: 07/23/2015] [Indexed: 12/15/2022] Open
Abstract
Most anti-angiogenic therapies currently being evaluated target the vascular endothelial growth factor (VEGF) pathway; however, the tumor vasculature can acquire resistance to VEGF-targeted therapy by shifting to other angiogenesis mechanisms. Therefore, other therapeutic agents that block non-VEGF angiogenic pathways need to be evaluated. Here, we identified ferulic acid as a novel fibroblast growth factor receptor 1 (FGFR1) inhibitor and a novel agent with potential anti-angiogenic and anti-cancer activities. Ferulic acid demonstrated inhibition of endothelial cell proliferation, migration and tube formation in response to basic fibroblast growth factor 1 (FGF1). In ex vivo and in vivo angiogenesis assays, ferulic acid suppressed FGF1-induced microvessel sprouting of rat aortic rings and angiogenesis. To understand the underlying molecular basis, we examined the effects of ferulic acid on different molecular components and found that ferulic acid suppressed FGF1-triggered activation of FGFR1 and phosphatidyl inositol 3-kinase (PI3K)-protein kinase B (Akt) signaling. Moreover, ferulic acid directly inhibited proliferation and blocked the PI3K-Akt pathway in melanoma cell. In vivo, using a melanoma xenograft model, ferulic acid showed growth-inhibitory activity associated with inhibition of angiogenesis. Taken together, our results indicate that ferulic acid targets the FGFR1-mediated PI3K-Akt signaling pathway, leading to the suppression of melanoma growth and angiogenesis.
Collapse
|