1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Deng F, Lei J, Qiu J, Zhao C, Wang X, Li M, Sun M, Zhang M, Gao Q. DNA methylation landscape in pregnancy-induced hypertension: progress and challenges. Reprod Biol Endocrinol 2024; 22:77. [PMID: 38978060 PMCID: PMC11229300 DOI: 10.1186/s12958-024-01248-0] [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: 03/10/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Gestational hypertension (PIH), especially pre-eclampsia (PE), is a common complication of pregnancy. This condition poses significant risks to the health of both the mother and the fetus. Emerging evidence suggests that epigenetic modifications, particularly DNA methylation, may play a role in initiating the earliest pathophysiology of PIH. This article describes the relationship between DNA methylation and placental trophoblast function, genes associated with the placental microenvironment, the placental vascular system, and maternal blood and vascular function, abnormalities of umbilical cord blood and vascular function in the onset and progression of PIH, as well as changes in DNA methylation in the progeny of PIH, in terms of maternal, fetal, and offspring. We also explore the latest research on DNA methylation-based early detection, diagnosis and potential therapeutic strategies for PIH. This will enable the field of DNA methylation research to continue to enhance our understanding of the epigenetic regulation of PIH genes and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Fengying Deng
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Jiahui Lei
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, 215153, P.R. China
| | - Chenxuan Zhao
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Xietong Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Min Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Miao Sun
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China.
| | - Meihua Zhang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| | - Qinqin Gao
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China.
| |
Collapse
|
3
|
Venou TM, Vetsiou E, Varelas C, Daniilidis A, Psarras K, Koravou EE, Koutra M, Touloumenidou T, Tsolakidis V, Papalexandri A, Minti F, Mandala E, Dinas K, Vlachaki E, Gavriilaki E. Increased Complement Activation and Decreased ADAMTS13 Activity Are Associated with Genetic Susceptibility in Patients with Preeclampsia/HELLP Syndrome Compared to Healthy Pregnancies: An Observational Case-Controlled Study. J Pers Med 2024; 14:387. [PMID: 38673014 PMCID: PMC11051193 DOI: 10.3390/jpm14040387] [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: 02/12/2024] [Revised: 03/13/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Preeclampsia is a progressive multi-systemic disorder characterized by proteinuria, critical organ damage, and new-onset hypertension. It can be further complicated by HELLP syndrome (hemolysis, elevated liver enzymes, low platelets), resulting in critical liver or renal damage, disseminated coagulation, and grand mal seizures. This study aimed to examine the involvement of ADAMTS13, von Willebrand, and the complement system in the pathogenesis of preeclampsia/HELLP syndrome. We studied 30 Caucasian preeclamptic pregnant women and a control group of 15 healthy pregnancies. Genetic sequencing of ADAMTS13 and complement regulatory genes (MiniSeq System, Illumina) was performed. The modified Ham test was used to check for complement activation, ADAMTS13 activity, von Willebrand antigen (vWFAg) levels, and soluble C5b-9 levels were measured. Patients with preeclampsia had a decreased ADAMTS13 activity and increased C5b-9 levels. The vWFAg was significantly correlated with ADAMTS13 activity (r = 0.497, p = 0.003). Risk-factor variants were found in the genes of ADAMTS13, C3, thrombomodulin, CFB, CFH, MBL2, and, finally, MASP2. A portion of pregnant women with preeclampsia showed a decline in ADAMTS13 activity, correlated with vWFAg levels. These patients also exhibited an elevated complement activation and high-risk genetic variants in regulatory genes. Further research is needed to determine if these factors can serve as reliable biomarkers.
Collapse
Affiliation(s)
- Theodora-Maria Venou
- Hematological Laboratory, 2nd Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece (E.V.); (V.T.)
| | - Evangelia Vetsiou
- Hematological Laboratory, 2nd Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece (E.V.); (V.T.)
| | - Christos Varelas
- Hematology Department, Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.V.); (E.-E.K.); (M.K.); (T.T.); (A.P.)
| | - Angelos Daniilidis
- 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Papageorgiou General Hospital, 56429 Thessaloniki, Greece;
| | - Kyriakos Psarras
- 2nd Propedeutical Department of Surgery, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Evaggelia-Evdoxia Koravou
- Hematology Department, Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.V.); (E.-E.K.); (M.K.); (T.T.); (A.P.)
| | - Maria Koutra
- Hematology Department, Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.V.); (E.-E.K.); (M.K.); (T.T.); (A.P.)
| | - Tasoula Touloumenidou
- Hematology Department, Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.V.); (E.-E.K.); (M.K.); (T.T.); (A.P.)
| | - Vasilis Tsolakidis
- Hematological Laboratory, 2nd Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece (E.V.); (V.T.)
| | - Apostolia Papalexandri
- Hematology Department, Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.V.); (E.-E.K.); (M.K.); (T.T.); (A.P.)
| | - Fani Minti
- Department of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Evdokia Mandala
- 4th Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Konstantinos Dinas
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Efthymia Vlachaki
- Hematological Laboratory, 2nd Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece (E.V.); (V.T.)
| | - Eleni Gavriilaki
- 2nd Propedeutical Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| |
Collapse
|
4
|
McCabe CF, Goodrich JM, Bakulski KM, Domino SE, Jones TR, Colacino J, Dolinoy DC, Padmanabhan V. Probing prenatal bisphenol exposures and tissue-specific DNA methylation responses in cord blood, cord tissue, and placenta. Reprod Toxicol 2023; 115:74-84. [PMID: 36473650 PMCID: PMC9851062 DOI: 10.1016/j.reprotox.2022.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
The early-gestational fetal epigenome establishes the landscape for fetal development and is susceptible to disruption via environmental stressors including chemical exposures. Research has explored how cell- and tissue-type-specific epigenomic signatures contribute to human disease, but how the epigenome in each tissue comparatively responds to environmental exposures is largely unknown. This pilot study compared DNA methylation in four previously identified genes across matched cord blood (CB), cord tissue (CT), and placental (PL) samples from 28 mother-infant pairs in tthe Michigan Mother Infant Pairs study; evaluated association between prenatal exposure to bisphenols (BPA, BPF, and BPS) and DNA methylation (DNAm) by tissue type; compared epigenome-wide DNAm of CB and PL; and explored associations between prenatal bisphenol exposures and epigenome-wide DNAm in PL. Bisphenol concentrations were quantified in first-trimester maternal urine. DNAm was assessed at four genes via pyrosequencing in three tissues; epigenome-wide DNAm analysis via Infinium MethylationEPIC array was completed on CB and PL. Candidate gene analysis revealed tissue-specific differences across all genes. In adjusted linear regression, BPA and BPF were associated with DNAm across candidate genes in PL but not CB and CT. Epigenome-wide comparison of matched CB and PL DNAm revealed tissue-specific differences at most CpG sites and modest associations between maternal first-trimester bisphenol exposures and PL but not CB DNAm. These data endorse inclusion of a variety of tissues in prenatal exposure studies. Overlapping and divergent responses in CB, CT, and PL demonstrate their utility in combination to capture a fuller picture of the epigenetic effect of developmental exposures.
Collapse
Affiliation(s)
- Carolyn F McCabe
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tamara R Jones
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Justin Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Qu H, Khalil RA. Role of ADAM and ADAMTS Disintegrin and Metalloproteinases in Normal Pregnancy and Preeclampsia. Biochem Pharmacol 2022; 206:115266. [PMID: 36191626 DOI: 10.1016/j.bcp.2022.115266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022]
Abstract
Normal pregnancy (NP) involves intricate processes starting with egg fertilization, proceeding to embryo implantation, placentation and gestation, and culminating in parturition. These pregnancy-related processes require marked uteroplacental and vascular remodeling by proteolytic enzymes and metalloproteinases. A disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) are members of the zinc-dependent family of proteinases with highly conserved protein structure and sequence homology, which include a pro-domain, and a metalloproteinase, disintegrin and cysteine-rich domain. In NP, ADAMs and ADAMTS regulate sperm-egg fusion, embryo implantation, trophoblast invasion, placental angiogenesis and spiral arteries remodeling through their ectodomain proteolysis of cell surface cytokines, cadherins and growth factors as well as their adhesion with integrins and cell-cell junction proteins. Preeclampsia (PE) is a serious complication of pregnancy characterized by new-onset hypertension (HTN) in pregnancy (HTN-Preg) at or after 20 weeks of gestation, with or without proteinuria. Insufficient trophoblast invasion of the uterine wall, inadequate expansive remodeling of the spiral arteries, reduced uteroplacental perfusion pressure, and placental ischemia/hypoxia are major initiating events in the pathogenesis of PE. Placental ischemia/hypoxia increase the release of reactive oxygen species (ROS), which lead to aberrant expression/activity of certain ADAMs and ADAMTS. In PE, abnormal expression/activity of specific ADAMs and ADAMTS that function as proteolytic sheddases could alter proangiogenic and growth factors, and promote the release of antiangiogenic factors and inflammatory cytokines into the placenta and maternal circulation leading to generalized inflammation, endothelial cell injury and HTN-Preg, renal injury and proteinuria, and further decreases in uteroplacental blood flow, exaggeration of placental ischemia, and consequently fetal growth restriction. Identifying the role of ADAMs and ADAMTS in NP and PE has led to a better understanding of the underlying molecular and vascular pathways, and advanced the potential for novel biomarkers for prediction and early detection, and new approaches for the management of PE.
Collapse
Affiliation(s)
- Hongmei Qu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA.
| |
Collapse
|
6
|
Timur B, Guney G. The role of serum ADAMTS-1 levels in Hyperemesis Gravidarum. BMC Pregnancy Childbirth 2022; 22:499. [PMID: 35725415 PMCID: PMC9208199 DOI: 10.1186/s12884-022-04832-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/10/2022] [Indexed: 11/19/2022] Open
Abstract
Background We aimed to investigate the levels of ADAMTS-1, which is secreted from the extracellular matrix during trophoblastic invasion in hyperemesis gravidarum (HEG). Methods In this cross-sectional study, we compared 45 HEG patients aged between 21 and 34 in terms of ADAMTS-1 levels with a control group consisting of 44 healthy pregnant women. The demographic characteristics and several laboratory parameters of the patients were recorded. Both groups were also compared in terms of ketonuria. We evaluated the correlation between ADAMTS-1 levels and ketonuria. Results The 2 groups were matched in terms of age, gestational age, gravidity, parity, and body mass index. Some inflammatory markers, such as neutrophil count, MPV, PDW, and PCT levels, were significantly higher in the HEG groups compared to the control group (all p < 0.05). However, mean MCV and serum TSH levels were statistically significantly lower in this group (both p < 0.001). ADAMTS-1 levels were 12.6 ± 1.4 ng/ml in the HEG group and 6.2 ± 1.6 ng/ml in the control group (p < 0.001). It was significantly and positively correlated with urine ketone, neutrophil count, and PDW, whereas negatively correlated with MCV and TSH value in the HEG group. ROC analysis showed that a threshold value of 11.275 ng/ml for ADAMTS-1 predicted HEG patients with a sensitivity of 60% and specificity of 95.5%. Conclusion ADAMTS-1 serum levels are increased in HEG patients, and there is a positive correlation between ADAMTS-1 levels and ketonuria.
Collapse
Affiliation(s)
- Burcu Timur
- Department of Obstetrics and Gynecology, Ordu University Training and Research Hospital, Bucak District, Nefsi Bucak Street, Ordu, 52200, Turkey.
| | - Gurhan Guney
- Department of Obstetrics and Gynecology, Balikesir University Medical Faculty, Balıkesir, Turkey
| |
Collapse
|
7
|
Molecular Advances in Preeclampsia and HELLP Syndrome. Int J Mol Sci 2022; 23:ijms23073851. [PMID: 35409211 PMCID: PMC8999044 DOI: 10.3390/ijms23073851] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Preeclampsia (PE) constitutes one of the principal reasons for maternal and perinatal morbidity and mortality worldwide. The circumstance typically implicates formerly healthful normotensive women, after 20 weeks of gestation, typically withinside the third trimester, without regarded threat elements or past deliveries. PE can be further complicated with hemolysis and thrombocytopenia, leading to the emergence of HELLP syndrome (Hemolysis, Elevated Liver enzymes, Low platelets). Both conditions are classified as hypertensive diseases of pregnancy (HDP), and their pathogenesis has been linked to an excessive maternal inflammatory response, accompanied by enhanced endothelial activation. Several studies have found that in pregnancies affected by PE/HELLP, von Willebrand factor (vWF) antigen levels (vWF:Ag) are significantly elevated, while its cleaving protease (ADAMTS-13, A Disintegrin-like and Metalloprotease with Thrombospondin type 1 motif, member 13) activity is normal to decreased. Furthermore, the higher urine excretion of the terminal complement complex C5b-9, as well as its greater deposition in the placental surface in preeclamptic women, imply that the utero-placental unit’s distinctive deficits are intimately tied to disproportionate complement activation. The goal of this updated evaluation is to provide the most up-to-date molecular advances in the pathophysiology of PE/HELLP syndromes. Recent medical data on vWF:Ag levels in patients with PE, ADAMTS-13, and dysregulation of the complement system, are highlighted and evaluated. Furthermore, we discuss the relationship between those entities and the progression of the disease, as well as their significance in the diagnostic process. Finally, considering the difficulties in analyzing and controlling those symptoms in pregnant women, we can provide a current diagnostic and therapeutic algorithm.
Collapse
|
8
|
Integrated bioinformatics analysis reveals novel key biomarkers and potential candidate small molecule drugs in gestational diabetes mellitus. Biosci Rep 2021; 41:228450. [PMID: 33890634 PMCID: PMC8145272 DOI: 10.1042/bsr20210617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is the metabolic disorder that appears during pregnancy. The current investigation aimed to identify central differentially expressed genes (DEGs) in GDM. The transcription profiling by array data (E-MTAB-6418) was obtained from the ArrayExpress database. The DEGs between GDM samples and non-GDM samples were analyzed. Functional enrichment analysis were performed using ToppGene. Then we constructed the protein–protein interaction (PPI) network of DEGs by the Search Tool for the Retrieval of Interacting Genes database (STRING) and module analysis was performed. Subsequently, we constructed the miRNA–hub gene network and TF–hub gene regulatory network. The validation of hub genes was performed through receiver operating characteristic curve (ROC). Finally, the candidate small molecules as potential drugs to treat GDM were predicted by using molecular docking. Through transcription profiling by array data, a total of 869 DEGs were detected including 439 up-regulated and 430 down-regulated genes. Functional enrichment analysis showed these DEGs were mainly enriched in reproduction, cell adhesion, cell surface interactions at the vascular wall and extracellular matrix organization. Ten genes, HSP90AA1, EGFR, RPS13, RBX1, PAK1, FYN, ABL1, SMAD3, STAT3 and PRKCA were associated with GDM, according to ROC analysis. Finally, the most significant small molecules were predicted based on molecular docking. This investigation identified hub genes, signal pathways and therapeutic agents, which might help us, enhance our understanding of the mechanisms of GDM and find some novel therapeutic agents for GDM.
Collapse
|
9
|
Mohamedi Y, Fontanil T, Cal S, Cobo T, Obaya ÁJ. ADAMTS-12: Functions and Challenges for a Complex Metalloprotease. Front Mol Biosci 2021; 8:686763. [PMID: 33996918 PMCID: PMC8119882 DOI: 10.3389/fmolb.2021.686763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Nineteen members of the ADAMTS family of secreted zinc metalloproteinases are present in the human degradome. A wide range of different functions are being attributed to these enzymes and the number of their known substrates is considerably increasing in recent years. ADAMTSs can participate in processes such as fertility, inflammation, arthritis, neuronal and behavioral disorders, as well as cancer. Since its first annotation in 2001, ADAMTS-12 has been described to participate in different processes displayed by members of this family of proteinases. In this sense, ADAMTS-12 performs essential roles in modulation and recovery from inflammatory processes such as colitis, endotoxic sepsis and pancreatitis. ADAMTS-12 has also been involved in cancer development acting either as a tumor suppressor or as a pro-tumoral agent. Furthermore, participation of ADAMTS-12 in arthritis or in neuronal disorders has also been suggested through degradation of components of the extracellular matrix. In addition, ADAMTS-12 proteinase activity can also be modified by interaction with other proteins and thus, can be an alternative way of modulating ADAMTS-12 functions. In this review we revised the most relevant findings about ADAMTS-12 function on the 20th anniversary of its identification.
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Departamento de Investigación, Instituto Ordóñez, Oviedo, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, Oviedo, Spain.,Instituto Asturiano de Odontología, Oviedo, Spain
| | - Álvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain.,Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
10
|
Marinello WP, Mohseni ZS, Cunningham SJ, Crute C, Huang R, Zhang JJ, Feng L. Perfluorobutane sulfonate exposure disrupted human placental cytotrophoblast cell proliferation and invasion involving in dysregulating preeclampsia related genes. FASEB J 2020; 34:14182-14199. [PMID: 32901980 DOI: 10.1096/fj.202000716rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
We reported that maternal PFBS, an emerging pollutant, exposure is positively associated with preeclampsia which can result from aberrant trophoblasts invasion and subsequent placental ischemia. In this study, we investigated the effects of PFBS on trophoblasts proliferation/invasion and signaling pathways. We exposed a human trophoblast line, HTR8/SVneo, to PFBS. Cell viability, proliferation, and cell cycle were evaluated by the MTS assay, Ki-67 staining, and flow cytometry, respectively. We assessed cell migration and invasion with live-cell imaging-based migration assay and matrigel invasion assay, respectively. Signaling pathways were examined by Western blot, RNA-seq, and qPCR. PFBS exposure interrupted cell proliferation and invasion in a dose-dependent manner. PFBS (100 μM) did not cause cell death but instead significant cell proliferation without cell cycle disruption. PFBS (10 and 100 μM) decreased cell migration and invasion, while PFBS (0.1 μM) significantly increased cell invasion but not migration. Further, RNA-seq analysis identified dysregulated HIF-1α target genes that are relevant to cell proliferation/invasion and preeclampsia, while Western Blot data showed the activation of HIF-1α, but not Notch, ERK1/2, (PI3K)AKT, and P38 pathways. PBFS exposure altered trophoblast cell proliferation/invasion which might be mediated by preeclampsia-related genes, suggesting a possible association between prenatal PFBS exposure and adverse placentation.
Collapse
Affiliation(s)
- William P Marinello
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Zahra S Mohseni
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Sarah J Cunningham
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Christine Crute
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Jun J Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Moore RM, Katri R, Kumar D, Mansour JM, Mercer B, Moore JJ. α-Lipoic acid blocks the GMCSF induced protease/protease inhibitor spectrum associated with fetal membrane weakening in-vitro. Placenta 2020; 97:79-88. [PMID: 32792069 DOI: 10.1016/j.placenta.2020.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We use an in-vitro human fetal membrane (FM) explant-based model to study inflammation-induced FM weakening, a prerequisite for PPROM. In this system, GMCSF is a critical intermediate, both necessary and sufficient for TNFα and thrombin induced FM weakening. α-Lipoic-acid (LA) blocks TNFα and thrombin, as well as GMCSF-induced weakening. Recently, we reported LA concomitantly blocks GMCSF-induction of MMPs 2, 9 and 10 and inhibition of TIMPs 1-3. The aim of this study was to show that LA blocks GMCSF-induced increases in additional proteases and reductions in additional protease inhibitors. METHODS FM fragments were cultured±LA and then±GMCSF. In other experiments, weak versus strong, fresh FM were cultured without additions. Fragments were strength tested and media analyzed by multiplex protein ELISA for proteases and protease inhibitors. RESULTS GMCSF induced FM weakening and concomitantly increased several Proteases (Cathepsin-S, Proteinase-3, Elastase-2) and decreased several protease inhibitors (NGAL, Cystatin-C, HE4 and Thrombospondin1). LA inhibited GMCSF-induced FM weakening and all enzymatic changes. Untreated weaker versus stronger regions of fresh FM showed comparable differences in proteases and protease inhibitor patterns to GMCSF-stimulated versus controls. CONCLUSION LA blocks GMCSF-induced human FM weakening and associated protease increases and inhibitor decreases. The GMCSF-induced spectrum of protease/protease-inhibitor changes is similar to that in the natural weak FM fragments. In concert with previously reported GMCSF-induced changes in MMPs & TIMPs, these other protease and protease-inhibitor changes presumably facilitate FM weakening and rupture. LA blocks these GMCSF effects and therefore may be a useful agent to prevent PPROM.
Collapse
Affiliation(s)
- R M Moore
- Department of Pediatrics, Case Western Reserve University, MetroHealth Medical Center, 44109, Cleveland, OH, USA
| | - R Katri
- Miami University, MetroHealth Medical Center, 44109, Oxford, OH, USA
| | - D Kumar
- Department of Pediatrics, Case Western Reserve University, MetroHealth Medical Center, 44109, Cleveland, OH, USA
| | - J M Mansour
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 100900 Euclid Ave, 44106, Cleveland, OH, USA
| | - B Mercer
- Department of Reproductive Biology, Case Western Reserve University, MetroHealth Medical Center, 44109, Cleveland, OH, USA
| | - J J Moore
- Department of Pediatrics, Case Western Reserve University, MetroHealth Medical Center, 44109, Cleveland, OH, USA; Department of Reproductive Biology, Case Western Reserve University, MetroHealth Medical Center, 44109, Cleveland, OH, USA.
| |
Collapse
|
12
|
Zhang L, Zhao F, Li C, Li H, Tang Q, Chen Y, Yao Y, Ding Z, Xu Y, Chen A, Liu S. Hypomethylation of DNA promoter upregulates ADAMTS7 and contributes to HTR-8/SVneo and JEG-3 cells abnormalities in pre-eclampsia. Placenta 2020; 93:26-33. [PMID: 32250736 DOI: 10.1016/j.placenta.2020.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Accumulating evidences have suggested a crucial role of epigenetics in the initiation and progression of pre-eclampsia (PE). Here, we studied the expression of the metalloproteinase ADAMTS7 and the methylation level of its promoter in PE placentas and investigated ADAMTS7 role in the pathogenesis of PE. METHODS We first explored ADAMTS7 expression in PE and normal placentas by reverse transcription quantitative PCR (RT-qPCR), western blot, and immunohistochemistry. Methylation specific PCR (MSP) and bisulfite sequencing PCR (BSP) were performed to evaluate the methylation status of ADAMTS7 promoter. Treatment with 5'-Aza was used to induce demethylation and thereby to explore the direct relationship between promoter methylation and ADAMTS7 expression. CCK8 assay, colony formation assay, and trans-well assay were conducted to assess the viability, migration, and invasion of HTR-8/SVneo and JEG-3 cells. RESULTS Our results showed that ADAMTS7 expression was upregulated in PE placentas. Methylation analysis revealed a hypomethylated status of ADAMTS7 promoter regions in PE placenta tissues. Besides, demethylation induced by 5'-Aza directly restored ADAMTS7 expression in trophoblast cells. Finally, overexpression of ADAMTS7 inhibited viability, migration, and invasion of HTR-8/SVneo and JEG-3 cells, while silence of ADAMTS7 by RNA interference reciprocally facilitated cell viability, migration and invasion in vitro. DISCUSSION Upregulation of ADAMTS7 by promoter hypomethylation in placenta might contribute to the etiology of PE via suppressing cell functions of trophoblasts.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China; Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Fei Zhao
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Chuan Li
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hong Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qian Tang
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China; Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yunqing Chen
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yushuang Yao
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zhaoxia Ding
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yinglei Xu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China; Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Aiping Chen
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China; Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
13
|
Mani S, Ghosh J, Lan Y, Senapati S, Ord T, Sapienza C, Coutifaris C, Mainigi M. Epigenetic changes in preterm birth placenta suggest a role for ADAMTS genes in spontaneous preterm birth. Hum Mol Genet 2019; 28:84-95. [PMID: 30239759 DOI: 10.1093/hmg/ddy325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/13/2018] [Indexed: 12/28/2022] Open
Abstract
Preterm birth (PTB) affects approximately 1 in 10 pregnancies and contributes to approximately 50% of neonatal mortality. However, despite decades of research, little is understood about the etiology of PTB, likely due to the multifactorial nature of the disease. In this study, we examined preterm and term placentas, from unassisted conceptions and those conceived using in vitro fertilization (IVF). IVF increases the risk of PTB and causes epigenetic change in the placenta and fetus; therefore, we utilized these patients as a unique population with a potential common etiology. We investigated genome-wide DNA methylation in placentas from term IVF, preterm IVF, term control (unassisted conception) and preterm control pregnancies and discovered epigenetic dysregulation of multiple genes involved in cell migration, including members of the ADAMTS family, ADAMTS12 and ADAMTS16. These genes function in extracellular matrix regulation and tumor cell invasion, processes replicated by invasive trophoblasts (extravillous trophoblasts (EVTs)) during early placentation. Though expression was similar between term and preterm placentas, we found that both genes demonstrate high expression in first- and second-trimester placenta, specifically in EVTs and syncytiotrophoblasts. When we knocked down ADAMTS12 or ADAMTS16in vitro, there was poor EVT invasion and reduced matrix metalloproteinase activity, reinforcing their critical role in placentation. In conclusion, utilizing a population at high risk for PTB, we have identified a role for ADAMTS gene methylation in regulating early placentation and susceptibility to PTB.
Collapse
Affiliation(s)
- Sneha Mani
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashri Ghosh
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, USA
| | - Yemin Lan
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Teri Ord
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Carmen Sapienza
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, USA
| | - Christos Coutifaris
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Melekoglu R, Yilmaz E, Ciftci O, Kafadar YT, Celik E. Associations between second-trimester amniotic fluid levels of ADAMTS4, ADAMTS5, IL-6, and TNF-α and spontaneous preterm delivery in singleton pregnancies. J Perinat Med 2019; 47:304-310. [PMID: 30730845 DOI: 10.1515/jpm-2018-0297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/14/2019] [Indexed: 11/15/2022]
Abstract
Background We investigated the roles of inflammatory cytokines and the A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family in the etiopathogenesis of spontaneous preterm delivery by comparing the ADAMTS4, ADAMTS5, interleukin (IL)-6, and tumor necrosis factor-α (TNF-α) levels in second-trimester amniotic fluid between pregnant women with preterm birth and term controls. Methods All pregnant women who underwent second-trimester amniocentesis for genetic analysis between January 1, 2016, and January 1, 2018, were enrolled in this study. From this cohort, 22 patients who subsequently experienced spontaneous preterm delivery before 34 weeks of pregnancy formed the study group, and 22 age- and body mass index (BMI)-matched patients without preterm birth constituted the control group. Results No significant differences were observed between the preterm birth and control groups in terms of age, BMI, obstetric history of preterm delivery, gestational age at amniocentesis, or indication for amniocentesis. The mean amniotic fluid levels of ADAMTS4 and ADAMTS5 were significantly increased in the preterm birth group compared to the control group (248.3±22.6 and 182.4±19.8 pg/mL, P=0.012; and 198.6±21.6 and 159.1±21.7 pg/mL, P=0.035, respectively). Significantly increased IL-6 and TNF-α levels were also detected in the amniotic fluid of women who experienced spontaneous preterm delivery, relative to controls (142.1±16.2 and 95.8±16.4 pg/mL, P<0.001; and 139.4±12.5 and 89.6±11.2 pg/mL, P<0.001, respectively). Conclusion The results of this study imply that increased mid-trimester amniotic fluid levels of ADAMTS4, ADAMTS5, IL-6, and TNF-α play an important role in the pathophysiology of spontaneous preterm delivery.
Collapse
Affiliation(s)
- Rauf Melekoglu
- Faculty of Medicine, Department of Obstetrics and Gynecology, University of Inonu, Malatya, Turkey
| | - Ercan Yilmaz
- Faculty of Medicine, Department of Obstetrics and Gynecology, University of Inonu, 44280 Malatya, Turkey, Tel.: +905369556180, Fax: +90422 3411217
| | - Osman Ciftci
- Faculty of Medicine, Department of Medical Pharmacology, University of Pamukkale, Denizli, Turkey
| | | | - Ebru Celik
- Faculty of Medicine, Department of Obstetrics and Gynecology, University of Koc, İstanbul, Turkey
| |
Collapse
|
15
|
Gutiérrez JA, Gómez I, Chiarello DI, Salsoso R, Klein AD, Guzmán-Gutiérrez E, Toledo F, Sobrevia L. Role of proteases in dysfunctional placental vascular remodelling in preeclampsia. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165448. [PMID: 30954558 DOI: 10.1016/j.bbadis.2019.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022]
Abstract
Preeclampsia is a syndrome characterised by vascular dysfunction, impaired angiogenesis, and hypertension during pregnancy. Even when the precise pathophysiology of preeclampsia remains elusive, impaired vascular remodelling and placental angiogenesis in the placental villi and defective trophoblast invasion of the uterus are proposed as crucial mechanisms in this syndrome. Reduced trophoblast invasion leads to reduced uteroplacental blood flow and oxygen availability and increased oxidative stress. These phenomena trigger the release of soluble factors into the maternal and foetoplacental circulation that are responsible of the clinical features of preeclampsia. New blood vessels generation as well as vascular remodelling are mechanisms that require expression and activity of different proteases, including matrix metalloproteases, a-disintegrin and metalloproteases, and a-disintegrin and metalloprotease with thrombospondin motifs. These proteases exert proteolysis of the extracellular matrix. Additionally, cathepsins, a family of proteolytic enzymes, are primarily located in lysosomes but are also released by cells to the extracellular space. This review focuses on the role that these proteases play in the regulation of the uterine trophoblast invasion and the placental vascular remodelling associated with preeclampsia.
Collapse
Affiliation(s)
- Jaime A Gutiérrez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago 7510157, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Isabel Gómez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago 7510157, Chile
| | - Delia I Chiarello
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Rocío Salsoso
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Andrés D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7590943, Chile
| | - Enrique Guzmán-Gutiérrez
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Luis Sobrevia
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, 4029, Queensland, Australia; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| |
Collapse
|