1
|
Murthi P, Kalionis B. Homeobox genes in the human placenta: Twists and turns on the path to find novel targets. Placenta 2024; 157:28-36. [PMID: 38908943 DOI: 10.1016/j.placenta.2024.06.010] [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: 03/16/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
Fetal growth restriction (FGR) is a clinically important human pregnancy disorder that is thought to originate early in pregnancy and while its aetiology is not well understood, the disorder is associated with placental insufficiency. Currently treatment for FGR is limited by increased surveillance using ultrasound monitoring and premature delivery, or corticosteroid medication in the third trimester to prolong pregnancy. There is a pressing need for novel strategies to detect and treat FGR at its early stage. Homeobox genes are well established as master regulators of early embryonic development and increasing evidence suggests they are also important in regulating early placental development. Most important is that specific homeobox genes are abnormally expressed in human FGR. This review focusses on identifying the molecular pathways controlled by homeobox genes in the normal and FGR-affected placenta. This information will begin to address the knowledge gap in the molecular aetiology of FGR and lay the foundation for identifying potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital and Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Victoria, Australia.
| | - Bill Kalionis
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital and Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
2
|
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
|
3
|
Xu X, Chen J, Zhao H, Pi Y, Lin G, Hu L. Single-Cell RNA-seq Analysis of a Human Embryonic Stem Cell to Endothelial Cell System Based on Transcription Factor Overexpression. Stem Cell Rev Rep 2023; 19:2497-2509. [PMID: 37537495 DOI: 10.1007/s12015-023-10598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Human embryonic stem cell (hESC)-derived endothelial cells (ECs) possess therapeutic potential in many diseases. Cytokine supplementation induction and transcription factor overexpression have become two mainstream methods of hESC-EC induction. Single-cell RNA-seq technology has been widely used to analyse dynamic processes during hESC-EC induction and components of induced endothelial cells. However, studies that used single-cell RNA-seq are mainly based on cytokine supplementation methods. In this study, we used a high-efficiency human embryonic stem cell-endothelial cell line (hESC-EC) called the "FLI1-PKC system" as a research model and employed single-cell RNA sequencing (scRNA-seq) to investigate the transcriptional landscape and cellular dynamics. METHODS The high-efficiency hESC-EC induction (FLI1-PKC) system was established in our previous study. We applied single-cell RNA sequencing (scRNA-seq) of the differentiated cells at different time points and investigated the gene expression profiles. RESULTS The FLI1-PKC induction system can directionally differentiate hESCs into mature endothelial cells with all the requisite functions. Unlike other hES-EC induction protocols, the FLI1-PKC method follows a different induction route; nonetheless, the transcriptome of induced endothelial cells (iECs) remains the same. The elevated number of activated transcription factors may explain why the FLI1-PKC system is more effective than other hES-EC protocols. CONCLUSION Our study has presented a single-cell transcriptional overview of a high-efficiency hESC-EC induction system, which can be used as a model and reference for further improvement in other hESC induction systems.
Collapse
Affiliation(s)
- XiangWang Xu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
| | - JunRu Chen
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China
| | - Hao Zhao
- Xiangya Hospital of Central South University, Changsha, Human, China
| | - YuZe Pi
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cell, Changsha, 410008, Hunan, China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cell, Changsha, 410008, Hunan, China.
| |
Collapse
|
4
|
Silini AR, Di Pietro R, Lang-Olip I, Alviano F, Banerjee A, Basile M, Borutinskaite V, Eissner G, Gellhaus A, Giebel B, Huang YC, Janev A, Kreft ME, Kupper N, Abadía-Molina AC, Olivares EG, Pandolfi A, Papait A, Pozzobon M, Ruiz-Ruiz C, Soritau O, Susman S, Szukiewicz D, Weidinger A, Wolbank S, Huppertz B, Parolini O. Perinatal Derivatives: Where Do We Stand? A Roadmap of the Human Placenta and Consensus for Tissue and Cell Nomenclature. Front Bioeng Biotechnol 2020; 8:610544. [PMID: 33392174 PMCID: PMC7773933 DOI: 10.3389/fbioe.2020.610544] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 02/05/2023] Open
Abstract
Progress in the understanding of the biology of perinatal tissues has contributed to the breakthrough revelation of the therapeutic effects of perinatal derivatives (PnD), namely birth-associated tissues, cells, and secreted factors. The significant knowledge acquired in the past two decades, along with the increasing interest in perinatal derivatives, fuels an urgent need for the precise identification of PnD and the establishment of updated consensus criteria policies for their characterization. The aim of this review is not to go into detail on preclinical or clinical trials, but rather we address specific issues that are relevant for the definition/characterization of perinatal cells, starting from an understanding of the development of the human placenta, its structure, and the different cell populations that can be isolated from the different perinatal tissues. We describe where the cells are located within the placenta and their cell morphology and phenotype. We also propose nomenclature for the cell populations and derivatives discussed herein. This review is a joint effort from the COST SPRINT Action (CA17116), which broadly aims at approaching consensus for different aspects of PnD research, such as providing inputs for future standards for the processing and in vitro characterization and clinical application of PnD.
Collapse
Affiliation(s)
- Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
| | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- StemTeCh Group, G. d’Annunzio Foundation, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Ingrid Lang-Olip
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - Asmita Banerjee
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mariangela Basile
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- StemTeCh Group, G. d’Annunzio Foundation, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Veronika Borutinskaite
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Günther Eissner
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Aleksandar Janev
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nadja Kupper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ana Clara Abadía-Molina
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Enrique G. Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Unidad de Gestión Clínica Laboratorios, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Assunta Pandolfi
- StemTeCh Group, G. d’Annunzio Foundation, G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Vascular and Stem Cell Biology, Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti-Pescara, CAST (Center for Advanced Studies and Technology, ex CeSI-MeT), Chieti, Italy
| | - Andrea Papait
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Department of Women’s and Children’s Health, University of Padova, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Olga Soritau
- The Oncology Institute “Prof. Dr. Ion Chiricuta”, Cluj-Napoca, Romania
| | - Sergiu Susman
- Department of Morphological Sciences-Histology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Pathology, IMOGEN Research Center, Cluj-Napoca, Romania
| | - Dariusz Szukiewicz
- Department of General and Experimental Pathology with Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
5
|
Alfaidy N, Brouillet S, Rajaraman G, Kalionis B, Hoffmann P, Barjat T, Benharouga M, Murthi P. The Emerging Role of the Prokineticins and Homeobox Genes in the Vascularization of the Placenta: Physiological and Pathological Aspects. Front Physiol 2020; 11:591850. [PMID: 33281622 PMCID: PMC7689260 DOI: 10.3389/fphys.2020.591850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/13/2020] [Indexed: 01/25/2023] Open
Abstract
Vasculogenesis and angiogenesis are key processes of placental development, which occur throughout pregnancy. Placental vasculogenesis occurs during the first trimester of pregnancy culminating in the formation of hemangioblasts from intra-villous stem cells. Placental angiogenesis occurs subsequently, forming new blood vessels from existing ones. Angiogenesis also takes place at the fetomaternal interface, allowing essential spiral arteriole remodeling to establish the fetomaternal circulation. Vasculogenesis and angiogenesis in animal models and in humans have been studied in a wide variety of in vitro, physiological and pathological conditions, with a focus on the pro- and anti-angiogenic factors that control these processes. Recent studies revealed roles for new families of proteins, including direct participants such as the prokineticin family, and regulators of these processes such as the homeobox genes. This review summarizes recent advances in understanding the molecular mechanisms of actions of these families of proteins. Over the past decade, evidence suggests increased production of placental anti-angiogenic factors, as well as angiogenic factors are associated with fetal growth restriction (FGR) and preeclampsia (PE): the most threatening pathologies of human pregnancy with systemic vascular dysfunction. This review also reports novel clinical strategies targeting members of these family of proteins to treat PE and its consequent effects on the maternal vascular system.
Collapse
Affiliation(s)
- Nadia Alfaidy
- Unité 1036, Institut National de la Santé et de la Recherche Médicale, Grenoble, France.,Department of Biology, University of Grenoble Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Sophie Brouillet
- INSERM U1203, Department of Reproductive Biology, University of Montpellier, Montpellier, France
| | - Gayathri Rajaraman
- Faculty of Health and Biomedicine, First Year College, Victoria University, St. Albans, VIC, Australia
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine, Obstetrics and Gynaecology, Pregnancy Research Centre, Royal Women's Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Pascale Hoffmann
- Unité 1036, Institut National de la Santé et de la Recherche Médicale, Grenoble, France.,Department of Biology, University of Grenoble Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Tiphaine Barjat
- Unité 1059, Saint-Etienne Hospital, Institut National de la Santé et de la Recherche Médicale, Saint-Étienne, France
| | - Mohamed Benharouga
- Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Padma Murthi
- Department of Maternal-Fetal Medicine, Obstetrics and Gynaecology, Pregnancy Research Centre, Royal Women's Hospital, The University of Melbourne, Parkville, VIC, Australia.,Department of Pharmacology, The Ritchie Centre, Monash Biomedicine Discovery Institute, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| |
Collapse
|
6
|
Cole A, Lee M, Koloff Z, Ghani KR. Complex Re-do robotic pyeloplasty using cryopreserved placental tissue: an adjunct for success. Int Braz J Urol 2020; 47:214-215. [PMID: 33047934 PMCID: PMC7712697 DOI: 10.1590/s1677-5538.ibju.2020.0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/04/2020] [Indexed: 11/22/2022] Open
Abstract
Introduction and Objectives: Management of recurrent ureteropelvic junction obstruction (UPJO) following pyeloplasty presents a challenging clinical problem. Failure of initial pyeloplasty is, in part, secondary to ureteral devascularization and subsequent fibrosis. In this video, we present a case of an anastomotic augmentation with cryopreserved placental tissue (CPT) to improve tissue healing and angiogenesis, and aid with the success of re-do robotic pyeloplasty. Materials and Methods: We present a 46-year-old female with history of recurrent left-sided UPJO treated by initial endopyelotomy and then open pyeloplasty. She underwent re-do robotic pyeloplasty (DaVinci Si™, Intuitive Surgical) with CPT. The patient was placed in the flank position; a 12mm camera port, three 8mm robotic ports, and a 12mm assistant port were used. The renal pelvis and upper ureter were mobilized to reveal a dense scar at the UPJ. A dismembered pyeloplasty was performed with barbed suture. After completion of the anastomosis, a section of CPT (Stravix™, Osiris Therapeutics) was wrapped around the anastomosis. CPT is composed of umbilical amnion and Wharton's jelly, which contains a mixture of extracellular matrix, and growth factors. The CPT is prepared and thawed on the bedside table, and placed into the peritoneum through the 12mm port in the correct orientation. The wrap is secured to the anastomosis with a fibrin sealant (EVICEL™, Johnson & Johnson). Results: The patient experienced resolution of flank pain. MAG3 renogram demonstrated resolution of obstruction at 6 months, with improvement of T½ time from 34 minutes to 7 minutes, with sustained improvement with repeat scan 18 months after surgery. Ureteroscopy demonstrated a patent UPJ. Strategies for successful robotic pyeloplasty after initial failed management include: (1) use of appropriate CPT agent to support the anastomosis - selection of thicker, more durable CPT to allow passage through laparoscopic port, (2) preparation on bedside table with enough time to allow thawing, (3) marking Wharton's jelly side of tissue for orientation, and (4) use of sealing agent to secure CPT to the anastomosis and prevent dislodgement. Conclusions: We demonstrate a novel approach to manage recurrent UPJ obstruction with robotic surgery using CPT. Placenta-derived products may have an increasing role in the performance of complex robotic urologic reconstructive surgery.
Collapse
Affiliation(s)
- Adam Cole
- Department of Urology, Michigan Medicine, Ann Arbor, MI, USA
| | - Matthew Lee
- Department of Urology, Michigan Medicine, Ann Arbor, MI, USA
| | - Zachary Koloff
- Department of Urology, Michigan Medicine, Ann Arbor, MI, USA
| | | |
Collapse
|
7
|
Zhou J, Guo X, Sun Y, Ma L, Zhe R. Levels of serum Hoxb3 and sFlt-1 in pre-eclamptic patients and their effects on pregnancy outcomes. J Obstet Gynaecol Res 2020; 46:2010-2018. [PMID: 32748508 DOI: 10.1111/jog.14397] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/21/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022]
Abstract
AIM We aimed to explore a new approach and theoretical basis for the diagnosis, treatment and prognosis of pre-eclampsia. METHODS In total, 103 pre-eclamptic patients (study group: SG) and 100 healthy pregnant women (control group: CG) were enrolled. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression level of serum Hoxb3. Enzyme-linked immunosorbent assay was used to detect the content of serum sFlt-1. Pregnancy outcomes in the two groups were recorded, and the correlations of the levels of Hoxb3 and sFlt-1 with the pregnancy outcomes were analyzed. RESULTS The relative expression of serum Hoxb3 mRNA in the CG was significantly higher than that in the SG, whereas the content of serum sFlt-1 in the CG was significantly lower than that in the SG. Compared with the CG, the SG had a significantly lower number of spontaneous deliveries, higher number of cesarean deliveries and significantly higher number of uneventful perinatal births. The incidences of intrauterine growth restriction, intrauterine distress, premature infants and neonatal deaths in perinatal infants in the SG were significantly higher than those in the CG. According to the analysis of receiver operating characteristic curves, the areas under the curves of Hoxb3, sFlt-1 and their combined detection for diagnosing pre-eclampsia were 0.799, 0.856 and 0.930, respectively. The areas under the curves for predicting poor perinatal outcomes were 0.724, 0.828 and 0.871, respectively. CONCLUSION In conclusion, Hoxb3 and sFlt-1 have certain reference significance for the risk evaluation of pre-eclampsia and the adverse pregnancy outcomes of pre-eclampsia women.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaohui Guo
- Department of Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| | - Yuan Sun
- Department of Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| | - Li Ma
- Department of Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| | - Ruilian Zhe
- Department of Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
8
|
Cai H, Zhu X, Li Z, Zhu Y, Lang J. lncRNA/mRNA profiling of endometriosis rat uterine tissues during the implantation window. Int J Mol Med 2019; 44:2145-2160. [PMID: 31638262 PMCID: PMC6844642 DOI: 10.3892/ijmm.2019.4370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023] Open
Abstract
Endometriosis is associated with changes in long non-coding RNA (lncRNA) and mRNA expression, but the exact changes during the implantation window are unknown. Therefore, this study aimed to explore the lncRNA and mRNA expression profiles in the uterus of rats with endometriosis during the implantation window. A total of 35 non-pregnant female rats were randomized to the endometriosis (n=13), adipose tissue control (n=8) and blank control (n=14) groups. On the 5th day of pregnancy, the rats were sacrificed to obtain uterine tissues. lncRNA and mRNA were analyzed using gene chips. A total of five differentially expressed lncRNA and four mRNA were validated by reverse transcription-quantitative (RT-q)PCR. Immunohistochemistry and western blotting were used to determine the expression of the ADAM metal-lopeptidase with thrombospondin type 1 motif 7 (Adamts7), tumor protein p53 (Tp53), distal-less homeobox 3 (Dlx3) and pyrimidinergic receptor P2Y6 (P2ry6) proteins. There were 115 upregulated lncRNAs, 51 downregulated lncRNAs, 97 upregulated mRNAs and 85 downregulated mRNAs in the endometriosis group. RT-qPCR confirmed the trends for five lncRNAs and four mRNAs (Adamts7, Tp53, Dlx3 and P2ry6). The relative protein expression levels of Adamts7, P2ry6, Dlx3 and TP53 were significantly different in the endometriosis group (P<0.05 vs. controls). Bioinformatics predicted the co-expression relationship of the selected five lncRNA and four mRNA. Gene ontology and the Kyoto Encyclopedia of Genes and Genomes predicted that Adamts7, P2ry6, Dlx3 and TP53 were involved in endometriosis-related inflammation and reproductive pathways. In conclusion, the changes in the expression of lncRNAs, mRNAs and proteins (Adamts7, P2ry6, Dlx3 and TP53) may possibly affect endometrial receptivity in rats with endometriosis during the implantation window, probably resulting in implantation failure of the embryo.
Collapse
Affiliation(s)
- Han Cai
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Zhanfei Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Yapei Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| |
Collapse
|
9
|
Liu Z, Guo Y, Lian F, Wang K, Sun Z, Wang Y, Zhang Z. Expression of HOXA10 gene in women with polycystic ovarian syndrome and its correlation analysis with lipid metabolism. MINERVA ENDOCRINOL 2019; 44:413-415. [PMID: 31347345 DOI: 10.23736/s0391-1977.19.03064-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Zhuo Liu
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Guo
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China -
| | - Fang Lian
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kehua Wang
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhengao Sun
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunhan Wang
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhumei Zhang
- Center for Reproduction and Genetics of Integrated Traditional Chinese and Western Medicine, Affiliate Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
10
|
Lu X, Cui J, Cui L, Luo Q, Cao Q, Yuan W, Zhang H. The effects of human umbilical cord-derived mesenchymal stem cell transplantation on endometrial receptivity are associated with Th1/Th2 balance change and uNK cell expression of uterine in autoimmune premature ovarian failure mice. Stem Cell Res Ther 2019; 10:214. [PMID: 31331391 PMCID: PMC6647296 DOI: 10.1186/s13287-019-1313-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023] Open
Abstract
Background To explore the mechanism of human umbilical cord-derived mesenchymal stem cell (hUMSC) transplantation to improve ovarian function and the endometrial receptivity in autoimmune premature ovarian failure (POF) mice. Methods The POF model was established in mice treated with zona pellucida 3 polypeptide fragment (zona pellucida 3, ZP3). The hUMSCs were transplanted into the POF mice through tail vein injection. Following the transplantation, the serum hormone levels of follicle stimulating hormone (FSH), estrogen (E2), progesterone (P), γ-interferon (IFN-γ), interleukin-2 (IL-2), and interleukin-4 (IL-4) were evaluated by ELISA analysis. Morphological changes of ovarian and uterus tissues were examined by HE staining and immunohistochemistry. The expression of Th1/Th2 cytokines of T cells in spleen and CD56+CD16− cells (uterine natural killer cells, uNK cells) in uterine was measured by flow cytometry (FCM) and immunohistochemistry. The expression of HOXA10 in uterine endometrium was examined by immunohistochemistry and RT-PCR analysis. The pinopodes of epithelial cells in uterine endometrium were examined by scanning electron microscopy. Results Following hUMSC transplantation, the serum levels of E2, P, and IL-4 were increased but FSH, IFN-γ, and IL-2 levels were decreased in POF mice. Also, the transplantation of hUMSCs caused an increase in total number of healthy follicles and decrease of atresia follicles. The expression of HOXA10 gene was significantly increased but the CD56+CD16− uNK cells decreased in the endometrium of uterine. The ratio of Th1/Th2 cytokines was also significantly decreased. Conclusion The data suggest that the recovery of ovarian function and endometrial receptivity in POF mice was regulated by the balance of Th1/Th2 cytokines and expression of uNK cells in the endometrium following hUMSC transplantation.
Collapse
Affiliation(s)
- Xueyan Lu
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.,College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Jingjing Cui
- The Affiliated Hospital of Binzhou Medical University, Binzhou, 256600, Shandong, China
| | - Linlu Cui
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.,College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Qianqian Luo
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.,College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Qizhi Cao
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Wendan Yuan
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.
| | - Hongqin Zhang
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China. .,College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China.
| |
Collapse
|
11
|
Gerbaud P, Murthi P, Guibourdenche J, Guimiot F, Sarazin B, Evain-Brion D, Badet J, Pidoux G. Study of Human T21 Placenta Suggests a Potential Role of Mesenchymal Spondin-2 in Placental Vascular Development. Endocrinology 2019; 160:684-698. [PMID: 30715257 DOI: 10.1210/en.2018-00826] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/25/2019] [Indexed: 12/12/2022]
Abstract
Placental development is particularly altered in trisomy of chromosome 21 (T21)-affected pregnancies. We previously described in T21-affected placentae an abnormal paracrine crosstalk between the villus mesenchymal core and villus trophoblasts. T21-affected placentae are known to be characterized by their hypovascularity. However, the causes of this anomaly remain not fully elucidated. Therefore, the hypothesis of an abnormal paracrine crosstalk between fetal mesenchymal core and placental endothelial cells (PLECs) was evocated. Villus mesenchymal cells from control (CMCs) and T21 placentae (T21MCs) were isolated and grown in culture to allow their characterization and collection of conditioned media for functional analyses (CMC-CM and T21MC-CM, respectively). Interestingly, PLEC proliferation and branching ability were less stimulated by T21MC-CM than by CMC-CM. Protein array analysis identified secreted proangiogenic growth factors in CMC-CM, which were reduced in T21MC-CM. Combined mass spectrometry and biochemical analysis identified spondin-2 as a factor decreased in T21MC-CM compared with CMC-CM. We found that exogenous spondin-2 stimulated PLEC proliferation and established that T21MC-CM supplemented with spondin-2 recovered conditioned media ability to induce PLEC proliferation and angiogenesis. Hence, this study demonstrates a crosstalk between villus mesenchymal and fetal endothelial cells, in which spondin-2 secreted from mesenchymal cells plays a central role in placental vascular functions. Furthermore, our results also suggest that a reduction in spondin-2 secretion may contribute to the pathogenesis of T21 placental hypovascularity.
Collapse
Affiliation(s)
- Pascale Gerbaud
- INSERM, UMR-S 1139, Paris, France
- INSERM, UMR-S 1180, Châtenay-Malabry, France
| | - Padma Murthi
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital, University of Melbourne, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jean Guibourdenche
- INSERM, UMR-S 1139, Paris, France
- Faculté de Pharmacie, Université Paris Descartes, Paris, France
- Service d'Hormonologie, Assistance Publique-Hôpitaux de Paris, CHU Cochin, Paris, France
- Fondation PremUP, Paris, France
| | - Fabien Guimiot
- Unité de Foetopathologie, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Paris, France
| | | | - Danièle Evain-Brion
- INSERM, UMR-S 1139, Paris, France
- Faculté de Pharmacie, Université Paris Descartes, Paris, France
- Fondation PremUP, Paris, France
| | - Josette Badet
- INSERM, UMR-S 1139, Paris, France
- Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Guillaume Pidoux
- INSERM, UMR-S 1139, Paris, France
- INSERM, UMR-S 1180, Châtenay-Malabry, France
| |
Collapse
|
12
|
Zhai J, Yao GD, Wang JY, Yang QL, Wu L, Chang ZY, Sun YP. Metformin Regulates Key MicroRNAs to Improve Endometrial Receptivity Through Increasing Implantation Marker Gene Expression in Patients with PCOS Undergoing IVF/ICSI. Reprod Sci 2019; 26:1439-1448. [PMID: 30599813 DOI: 10.1177/1933719118820466] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To some extent, the use of metformin may improve endometrial receptivity and pregnancy outcomes of women with polycystic ovarian syndrome (PCOS) undergoing in vitro fertilization/intracytoplasmic sperm injection. However, the mechanism is not well-known. The endometrium of metformin-treated group (metformin-treated patients with PCOS) and the control group (non-metformin-treated patients with PCOS) were analyzed for the expression of homeobox A10 (HOXA10) and integrin beta-3 (ITGB3) and differential micro RNA (miRNA) expression profiles. On this basis, miRDB and Target Scan databases were used to predict and screen out that miR-491-3p and miR-1910-3p may target HOXA10 and ITGB3. Furthermore, we verified the effects of metformin on the expression of HOXA10 and ITGB3, and regulatory effects of miR-1910-3p and miR-491-3p on HOXA10 and ITGB3 using Ishikawa cell line. Metformin induced a significant dose-dependent upregulation of HOXA10 and ITGB3. The results from the microarray analyses showed there were 40 differentially expressed miRNAs between the 2 groups. Among them, miR-1910-3p and miR-491-3p were the 2 significantly downregulated miRNAs. Bioinformatics prediction indicated that HOXA10 and ITGB3 are potential target genes for miR-1910-3p and miR-491-3p. In Ishikawa cells transfected with miR-491-3p mimics, the expression of HOXA10 and ITGB3 on both messenger RNA (mRNA) and protein level were lower than those in control group (P < .001). Also, the expression of HOXA10 mRNA and protein was lower in Ishikawa cells transfected with miR-1910-3p mimics (P < .001). However, no significant changes in ITGB3 levels were observed in cells transfected with miR-1910-3p mimics (P > .05). Metformin likely improves endometrial receptivity through downregulating the expression of miR-491-3p and miR-1910-3p, thereby increasing the expression of HOXA10 and ITGB3 in the endometrium of PCOS women.
Collapse
Affiliation(s)
- Jun Zhai
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Gui-Dong Yao
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jing-Yuan Wang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Qing-Ling Yang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Liang Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zi-Yin Chang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ying-Pu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
13
|
Diversity in human placental microvascular endothelial cells and macrovascular endothelial cells. Cytokine 2018; 111:287-294. [PMID: 30269024 DOI: 10.1016/j.cyto.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 02/02/2023]
Abstract
Angiogenesis is fundamental to normal placental development, and aberrant angiogenesis contributes substantially to placental pathologies. Placental angiogenesis is a pivotal process that plays a key mechanistic role in the elaboration of the placental villous tree, which is mainly taken by human placental microvascular endothelial cells (HPMECs), present in the fetal capillaries of chorionic villi, and macrovascular human umbilical vein endothelial cells (HUVECs) also play a role in this process. These are the two types of endothelial cells that form the placenta and differ in morphology and function. The placental vasculature represents a distinct territory that is highly specialized in structure and function. To distinguish the differences between HPMECs and HUVECs, we isolated HPMECs by paramagnetic particle separation and HUVECs through trypsinization and validated their characteristics. Then, we examined their response to fibroblast growth factor 2 (FGF2), vascular endothelial growth factor (VEGF) and endocrine-gland-derived vascular endothelial growth factor (EG-VEGF), as well as the underlying signaling mechanisms and their transcriptomes. We found that cultured HPMECs and HUVECs took up DiI-Ac-LDL and formed capillary-like tube structures on Matrigel. HPMECs and HUVECs had different expressions of eNOS, PROKR1 and PROKR2, and these characteristics substantiate the endothelial nature of cultured cells. FGF2 and VEGF stimulated the proliferation and migration of HPMECs and HUVECs via activation of PI3K/AKT1 and MEK1/MEK2/ERK1/ERK2. Interestingly, EG-VEGF increased the proliferation and migration of HPMECs via only MEK1/MEK2/ERK1/ERK2 and not PI3K/AKT1. Microarray analysis showed that there were some differentially expressed genes between HPMECs and HUVECs. Gene ontology analysis indicated that the differentially expressed genes were highly related to G-protein coupled receptor signaling pathway, angiogenesis, L-lysine transmembrane transport and blood vessel remodeling. These data provided evidence of heterogeneity between microvascular HPMECs and macrovascular HUVECs that most likely reflected significant differences in endothelial cell function in the two different cellular environments.
Collapse
|
14
|
Expression of Homeobox Gene HLX and its Downstream Target Genes are Altered in Placentae From Discordant Twin Pregnancies. Twin Res Hum Genet 2017; 21:42-50. [PMID: 29212571 DOI: 10.1017/thg.2017.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A discordant twin gestation, in which one fetus is significantly growth restricted, compared to the other normal twin, is a unique model that can be used to elucidate the mechanism(s) by which the intrauterine environment affects fetal growth. In many model systems, placental transcription factor genes regulate fetal growth. Transcription factors regulate growth through their activation or repression of downstream target genes that mediate important cell functions. The objective of this study was to determine the expression of the placental HLX homeobox gene transcription factor and its downstream target genes in dizygotic twins with growth discordance. In this cross-sectional study, HLX and its downstream target genes' retinoblastoma 1 (RB1) and cyclin kinase D (CDKN1C) expression levels were determined in placentae obtained from dichorionic diamniotic twin pregnancies (n = 23) where one of the twins was growth restricted. Fetal growth restriction (FGR) was defined as small for gestational age with abnormal umbilical artery Doppler indices when compared with the normal control co-twin. Homeobox gene HLX expression was significantly decreased at both the mRNA and protein levels in FGR twin placentae compared with the normal control co-twin placentae (p < .05). Downstream target genes CDKN1C and RB1 were also significantly decreased and increased, respectively, at both the mRNA and protein levels in FGR twin placentae compared with normal control co-twin placentae (p < .05). Together, these observations suggest an important association between HLX transcription factor expression and abnormal human placental development in discordant twin pregnancies.
Collapse
|
15
|
Chen H, Song Y, Yang S, Fu J, Feng X, Huang W. YAP mediates human decidualization of the uterine endometrial stromal cells. Placenta 2017; 53:30-35. [PMID: 28487017 DOI: 10.1016/j.placenta.2017.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 01/19/2023]
Abstract
INTRODUCTION The decidualization of uterine endometrial stromal cells (ESCs) is critical for the successful establishment and maintenance of pregnancy and involves extensive cell proliferation and differentiation. A newly established signaling pathway, the Hippo/Yes-associated protein (YAP) pathway, plays a critical role in these proliferation processes. Our previous study demonstrated that YAP is expressed in human ESCs. However, its role in decidualization remains unclear. The objective of the present study was to explore the role of YAP in the decidualization of human ESCs. METHODS The expression of YAP was first investigated in the endometrium of non-pregnant women and in the decidua of pregnant women. The role of YAP was investigated by transfecting ESCs with mRNA silencing constructs and observing the negative effects of this action upon decidualization induced in vitro. RESULTS Our results revealed that the expression of YAP was higher in decidual cells from early pregnant decidua compared with ESCs from non-pregnant endometrium. The expression levels of YAP and TEA domain 1 (TEAD1) were both increased in ESCs during in vitro decidualization and the knockdown of YAP in ESCs caused negative effects upon decidualization in vitro. DISCUSSION Our study suggests that YAP is upregulated in human decidual cells compared with ESCs and influences the decidualization of ESCs.
Collapse
Affiliation(s)
- Hengxi Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Yong Song
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Shiyuan Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Jing Fu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Xue Feng
- Department of Obstetrics and Gynecology, Chongqing Obstetrics and Gynecology Hospital, Chongqing, People's Republic of China
| | - Wei Huang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
16
|
Elçin AE, Parmaksiz M, Dogan A, Seker S, Durkut S, Dalva K, Elçin YM. Differential gene expression profiling of human adipose stem cells differentiating into smooth muscle-like cells by TGFβ1/BMP4. Exp Cell Res 2017; 352:207-217. [DOI: 10.1016/j.yexcr.2017.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/18/2022]
|
17
|
Liu Z, Ji Z, Wang G, Chao T, Hou L, Wang J. Genome-wide analysis reveals signatures of selection for important traits in domestic sheep from different ecoregions. BMC Genomics 2016; 17:863. [PMID: 27809776 PMCID: PMC5094087 DOI: 10.1186/s12864-016-3212-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 10/25/2016] [Indexed: 12/22/2022] Open
Abstract
Background Throughout a long period of adaptation and selection, sheep have thrived in a diverse range of ecological environments. Mongolian sheep is the common ancestor of the Chinese short fat-tailed sheep. Migration to different ecoregions leads to changes in selection pressures and results in microevolution. Mongolian sheep and its subspecies differ in a number of important traits, especially reproductive traits. Genome-wide intraspecific variation is required to dissect the genetic basis of these traits. Results This research resequenced 3 short fat-tailed sheep breeds with a 43.2-fold coverage of the sheep genome. We report more than 17 million single nucleotide polymorphisms and 2.9 million indels and identify 143 genomic regions with reduced pooled heterozygosity or increased genetic distance to each other breed that represent likely targets for selection during the migration. These regions harbor genes related to developmental processes, cellular processes, multicellular organismal processes, biological regulation, metabolic processes, reproduction, localization, growth and various components of the stress responses. Furthermore, we examined the haplotype diversity of 3 genomic regions involved in reproduction and found significant differences in TSHR and PRL gene regions among 8 sheep breeds. Conclusions Our results provide useful genomic information for identifying genes or causal mutations associated with important economic traits in sheep and for understanding the genetic basis of adaptation to different ecological environments. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3212-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhaohua Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Zhibin Ji
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Guizhi Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Lei Hou
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China.
| |
Collapse
|
18
|
Gunatillake T, Yong HEJ, Dunk CE, Keogh RJ, Borg AJ, Cartwright JE, Whitley GS, Murthi P. Homeobox gene TGIF-1 is increased in placental endothelial cells of human fetal growth restriction. Reproduction 2016; 152:457-65. [PMID: 27539603 DOI: 10.1530/rep-16-0068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/18/2016] [Indexed: 01/09/2023]
Abstract
Aberrant placental angiogenesis is associated with fetal growth restriction (FGR). In mice, targeted disruption of the homeobox gene, transforming growth β-induced factor (Tgif-1), which is also a transcription factor, causes defective placental vascularisation. Nevertheless, the role of TGIF-1 in human placental angiogenesis is unclear. We have previously reported increased TGIF-1 expression in human FGR placentae and demonstrated localisation of TGIF-1 protein in placental endothelial cells (ECs). However, its functional role remains to be investigated. In this study, we aimed to specifically compare TGIF-1 mRNA expression in placental ECs isolated from human FGR-affected pregnancies with gestation-matched control pregnancies in two independent cohorts from Australia and Canada and to identify the functional role of TGIF-1 in placental angiogenesis using the human umbilical vein endothelial cell-derived cell line, SGHEC-7, and primary human umbilical vein ECs. Real-time PCR revealed that TGIF-1 mRNA expression was significantly increased in ECs isolated from FGR-affected placentae compared with that of controls. The functional roles of TGIF-1 were determined in ECs after TGIF-1 siRNA transfection. TGIF-1 inactivation in ECs significantly reduced TGIF-1 at both the mRNA and protein levels, as well as the proliferative and invasive potential, but significantly increased the angiogenic potential. Using angiogenesis PCR screening arrays, we identified ITGAV, NRP-1, ANPGT-1 and ANPGT-2 as novel downstream targets of TGIF-1, after TGIF-1 inactivation in ECs. Collectively, these results show that TGIF-1 regulates EC function and the expression of angiogenic molecules; and when abnormally expressed, may contribute to the aberrant placental angiogenesis observed in FGR.
Collapse
Affiliation(s)
- Tilini Gunatillake
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Hannah E J Yong
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Caroline E Dunk
- Lunenfeld Tanenbaum-Research InstituteMount Sinai Hospital, Toronto, Ontario, Canada
| | - Rosemary J Keogh
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Borg
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia
| | - Judith E Cartwright
- Institute of Cardiovascular and Cell SciencesSt George's, University of London, London, UK
| | - Guy S Whitley
- Institute of Cardiovascular and Cell SciencesSt George's, University of London, London, UK
| | - Padma Murthi
- Department of Maternal-Fetal Medicine Pregnancy Research CentreThe Royal Women's Hospital, Parkville, Victoria, Australia Department of Obstetrics and GynaecologyThe University of Melbourne, Parkville, Victoria, Australia Department of MedicineSchool of Clinical Sciences, Monash University, Clayton, Victoria, Australia The Ritchie CentreHudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
19
|
Slany A, Bileck A, Kreutz D, Mayer RL, Muqaku B, Gerner C. Contribution of Human Fibroblasts and Endothelial Cells to the Hallmarks of Inflammation as Determined by Proteome Profiling. Mol Cell Proteomics 2016; 15:1982-97. [PMID: 27025457 DOI: 10.1074/mcp.m116.058099] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
In order to systematically analyze proteins fulfilling effector functionalities during inflammation, here we present a comprehensive proteome study of inflammatory activated primary human endothelial cells and fibroblasts. Cells were stimulated with interleukin 1-β and fractionated in order to obtain secreted, cytoplasmic and nuclear protein fractions. Proteins were submitted to a data-dependent bottom up analytical platform using a QExactive orbitrap and the MaxQuant software for protein identification and label-free quantification. Results were further combined with similarly generated data previously obtained from the analysis of inflammatory activated peripheral blood mononuclear cells. Applying a false discovery rate of less than 0.01 at both, peptide and protein level, a total of 8370 protein groups assembled from 117,599 peptides was identified; mass spectrometry data have been made fully accessible via ProteomeXchange with identifier PXD003406 to PXD003417.Comparative proteome analysis allowed us to determine common and cell type-specific inflammation signatures comprising novel candidate marker molecules and related expression patterns of transcription factors. Cardinal features of inflammation such as interleukin 1-β processing and the interferon response differed substantially between the investigated cells. Furthermore, cells also exerted similar inflammation-related tasks; however, by making use of different sets of proteins. Hallmarks of inflammation thus emerged, including angiogenesis, extracellular matrix reorganization, adaptive and innate immune responses, oxidative stress response, cell proliferation and differentiation, cell adhesion and migration in addition to monosaccharide metabolic processes, representing both, common and cell type-specific responsibilities of cells during inflammation.
Collapse
Affiliation(s)
- Astrid Slany
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| | - Andrea Bileck
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| | - Dominique Kreutz
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| | - Rupert L Mayer
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| | - Besnik Muqaku
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| | - Christopher Gerner
- From the ‡Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Austria
| |
Collapse
|
20
|
Dakouane-Giudicelli M, Brouillet S, Traboulsi W, Torre A, Vallat G, Si Nacer S, Vallée M, Feige JJ, Alfaidy N, de Mazancourt P. Inhibition of human placental endothelial cell proliferation and angiogenesis by netrin-4. Placenta 2015; 36:1260-5. [PMID: 26390805 DOI: 10.1016/j.placenta.2015.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Netrin-4 is a secreted member of the laminin-related protein family, known to be involved in axonal guidance and endothelial cell survival, proliferation, and migration. We have recently reported the cellular localization of netrin-4 and its receptor neogenin in human first trimester and term placenta. A strong expression of netrin-4 was observed in trophoblast and in endothelial cells, suggesting a potential role of this protein in placental angiogenesis. In relation to human pregnancy, it has been reported that circulating netrin-4 were increased in fetal umbilical cord blood of intrauterine growth restriction IUGR compared to normal pregnancy suggesting an adverse effect of this protein on placental and fetal development. The aim of this study was to determine the role of netrin-4 in placental angiogenesis. METHODS The effects of netrin-4 on proliferation, migration, tube-like organization, and spheroid sprouting of human placental microvascular endothelial cells (HPEC) were studied. RESULTS We demonstrated that netrin-4 inhibits HPEC proliferation, tube-like formation, migration and spheroid sprouting, suggesting a direct role of netrin-4 in the regulation of intra-villus angiogenesis. DISCUSSION This is the first report of an anti-angiogenic activity of netrin-4 in human placenta. This study brings new insights into netrin-4 roles in placental angiogenesis and suggests possible involvements of netrin-4 in angiogenesis-related pathologies such as IUGR.
Collapse
Affiliation(s)
- M Dakouane-Giudicelli
- Institut National de la Santé et de la Recherche Médicale, Unité 1179, Montigny Le Bretonneux, France; Université de Versailles Saint Quentin, En Yvelines, France.
| | - S Brouillet
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France; Université Grenoble-Alpes, 38000 Grenoble, France; Commissariat à l'Energie Atomique (CEA), iRTSV-Biology of Cancer and Infection, Grenoble, France; Centre Hospitalier Universitaire de Grenoble, Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation, 38700 La Tronche, France
| | - W Traboulsi
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France
| | - A Torre
- Université de Versailles Saint Quentin, En Yvelines, France
| | - G Vallat
- Université de Versailles Saint Quentin, En Yvelines, France
| | - S Si Nacer
- Université de Versailles Saint Quentin, En Yvelines, France
| | - M Vallée
- Université de Versailles Saint Quentin, En Yvelines, France
| | - J J Feige
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France; Université Grenoble-Alpes, 38000 Grenoble, France; Commissariat à l'Energie Atomique (CEA), iRTSV-Biology of Cancer and Infection, Grenoble, France
| | - N Alfaidy
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France; Université Grenoble-Alpes, 38000 Grenoble, France; Commissariat à l'Energie Atomique (CEA), iRTSV-Biology of Cancer and Infection, Grenoble, France
| | - P de Mazancourt
- Institut National de la Santé et de la Recherche Médicale, Unité 1179, Montigny Le Bretonneux, France; Université de Versailles Saint Quentin, En Yvelines, France; AP-HP Hopital Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
21
|
Ren JG, Chen G, Zhu JY, Zhang W, Sun YF, Jia J, Zhang J, Zhao YF. Downregulation of the transforming growth factor-β/connective tissue growth factor 2 signalling pathway in venous malformations: its target potential for sclerotherapy. Br J Dermatol 2014; 171:242-51. [PMID: 24655310 DOI: 10.1111/bjd.12977] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2014] [Indexed: 01/04/2023]
Affiliation(s)
- J.-G. Ren
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - G. Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - J.-Y. Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - W. Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - Y.-F. Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - J. Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - J. Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - Y.-F. Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School and Hospital of Stomatology; Wuhan University; Wuhan China
| |
Collapse
|
22
|
Murthi P, Abumaree M, Kalionis B. Analysis of homeobox gene action may reveal novel angiogenic pathways in normal placental vasculature and in clinical pregnancy disorders associated with abnormal placental angiogenesis. Front Pharmacol 2014; 5:133. [PMID: 24926269 PMCID: PMC4045154 DOI: 10.3389/fphar.2014.00133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/14/2014] [Indexed: 11/13/2022] Open
Abstract
Homeobox genes are essential for both the development of the blood and lymphatic vascular systems, as well as for their maintenance in the adult. Homeobox genes comprise an important family of transcription factors, which are characterized by a well conserved DNA binding motif; the homeodomain. The specificity of the homeodomain allows the transcription factor to bind to the promoter regions of batteries of target genes and thereby regulates their expression. Target genes identified for homeodomain proteins have been shown to control fundamental cell processes such as proliferation, differentiation, and apoptosis. We and others have reported that homeobox genes are expressed in the placental vasculature, but our knowledge of their downstream target genes is limited. This review highlights the importance of studying the cellular and molecular mechanisms by which homeobox genes and their downstream targets may regulate important vascular cellular processes such as proliferation, migration, and endothelial tube formation, which are essential for placental vasculogenesis and angiogenesis. A better understanding of the molecular targets of homeobox genes may lead to new therapies for aberrant angiogenesis associated with clinically important pregnancy pathologies, including fetal growth restriction and preeclampsia.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital Parkville, VIC, Australia ; Department of Obstetrics and Gynaecology, The University of Melbourne Parkville, VIC, Australia ; NorthWest Academic Centre, The University of Melbourne St. Albans, VIC, Australia
| | - Mohamed Abumaree
- College of Science and Health Professions, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences Riyadh, Saudi Arabia
| | - Bill Kalionis
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital Parkville, VIC, Australia ; Department of Obstetrics and Gynaecology, The University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
23
|
Liu H, Murthi P, Qin S, Kusuma GD, Borg AJ, Knöfler M, Haslinger P, Manuelpillai U, Pertile MD, Abumaree M, Kalionis B. A novel combination of homeobox genes is expressed in mesenchymal chorionic stem/stromal cells in first trimester and term pregnancies. Reprod Sci 2014; 21:1382-94. [PMID: 24692208 DOI: 10.1177/1933719114526471] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human chorionic mesenchymal stem/stromal cells (CMSCs) derived from the placenta are similar to adult tissue-derived MSCs. The aim of this study was to investigate the role of these cells in normal placental development. Transcription factors, particularly members of the homeobox gene family, play crucial roles in maintaining stem cell proliferation and lineage specification in embryonic tissues. In adult tissues and organs, stem cells proliferate at low levels in their niche until they receive cues from the microenvironment to differentiate. The homeobox genes that are expressed in the CMSC niche in placental tissues have not been identified. We used the novel strategy of laser capture microdissection to isolate the stromal component of first trimester villi and excluded the cytotrophoblast and syncytiotrophoblast layers that comprise the outer layer of the chorionic villi. Microarray analysis was then used to screen for homeobox genes in the microdissected tissue. Candidate homeobox genes were selected for further RNA analysis. Immunohistochemistry of candidate genes in first trimester placental villous stromal tissue revealed homeobox genes Meis1, myeloid ectropic viral integration site 1 homolog 2 (MEIS2), H2.0-like Drosophila (HLX), transforming growth factor β-induced factor (TGIF), and distal-less homeobox 5 (DLX5) were expressed in the vascular niche where CMSCs have been shown to reside. Expression of MEIS2, HLX, TGIF, and DLX5 was also detected in scattered stromal cells. Real-time polymerase chain reaction and immunocytochemistry verified expression of MEIS2, HLX, TGIF, and DLX5 homeobox genes in first trimester and term CMSCs. These data suggest a combination of regulatory homeobox genes is expressed in CMSCs from early placental development to term, which may be required for stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Haiying Liu
- Department of Obstetrics and Gynaecology, QiLu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Padma Murthi
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Sharon Qin
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Gina D Kusuma
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Anthony J Borg
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Martin Knöfler
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Haslinger
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Ursula Manuelpillai
- Centre for Genetic Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria
| | - Mark D Pertile
- VCGS, Murdoch Children's Research Institute, Royal Childrens Hospital, Flemington Road, Parkville, Victoria, Australia
| | - Mohamed Abumaree
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences/ King Abdulla International Medical Research Center, Riyadh, Saudi Arabia
| | - Bill Kalionis
- Department of Obstetrics and Gynaecology, University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
24
|
Casanello P, Schneider D, Herrera EA, Uauy R, Krause BJ. Endothelial heterogeneity in the umbilico-placental unit: DNA methylation as an innuendo of epigenetic diversity. Front Pharmacol 2014; 5:49. [PMID: 24723887 PMCID: PMC3973902 DOI: 10.3389/fphar.2014.00049] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 03/06/2014] [Indexed: 12/20/2022] Open
Abstract
The endothelium is a multifunctional heterogeneous tissue playing a key role in the physiology of every organ. To accomplish this role the endothelium presents a phenotypic diversity that is early prompted during vascular development, allowing it to cope with specific requirements in a time- and site-specific manner. During the last decade several reports show that endothelial diversity is also present in the umbilico-placental vasculature, with differences between macro- and microvascular vessels as well as arterial and venous endothelium. This diversity is evidenced in vitro as a higher angiogenic capacity in the microcirculation; or disparity in the levels of several molecules that control endothelial function (i.e., receptor for growth factors, vasoactive mediators, and adhesion molecules) which frequently are differentially expressed between arterial and venous endothelium. Emerging evidence suggests that endothelial diversity would be prominently driven by epigenetic mechanisms which also control the basal expression of endothelial-specific genes. This review outlines evidence for endothelial diversity since early stages of vascular development and how this heterogeneity is expressed in the umbilico-placental vasculature. Furthermore a brief picture of epigenetic mechanisms and their role on endothelial physiology emphasizing new data on umbilical and placental endothelial cells is presented. Unraveling the role of epigenetic mechanisms on long term endothelial physiology and its functional diversity would contribute to develop more accurate therapeutic interventions. Altogether these data show that micro- versus macro-vascular, or artery versus vein comparisons are an oversimplification of the complexity occurring in the endothelium at different levels, and the necessity for the future research to establish the precise source of cells which are under study.
Collapse
Affiliation(s)
- Paola Casanello
- Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile ; Division of Paediatrics, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Daniela Schneider
- Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Emilio A Herrera
- Programa de Fisiopatologïa, Laboratorio de Función y Reactividad Vascular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile Santiago, Chile
| | - Ricardo Uauy
- Division of Paediatrics, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Bernardo J Krause
- Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
25
|
Toshner M, Dunmore BJ, McKinney EF, Southwood M, Caruso P, Upton PD, Waters JP, Ormiston ML, Skepper JN, Nash G, Rana AA, Morrell NW. Transcript analysis reveals a specific HOX signature associated with positional identity of human endothelial cells. PLoS One 2014; 9:e91334. [PMID: 24651450 PMCID: PMC3961275 DOI: 10.1371/journal.pone.0091334] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/10/2014] [Indexed: 01/16/2023] Open
Abstract
The endothelial cell has a remarkable ability for sub-specialisation, adapted to the needs of a variety of vascular beds. The role of developmental programming versus the tissue contextual environment for this specialization is not well understood. Here we describe a hierarchy of expression of HOX genes associated with endothelial cell origin and location. In initial microarray studies, differential gene expression was examined in two endothelial cell lines: blood derived outgrowth endothelial cells (BOECs) and pulmonary artery endothelial cells. This suggested shared and differential patterns of HOX gene expression between the two endothelial lines. For example, this included a cluster on chromosome 2 of HOXD1, HOXD3, HOXD4, HOXD8 and HOXD9 that was expressed at a higher level in BOECs. Quantative PCR confirmed the higher expression of these HOXs in BOECs, a pattern that was shared by a variety of microvascular endothelial cell lines. Subsequently, we analysed publically available microarrays from a variety of adult cell and tissue types using the whole “HOX transcriptome” of all 39 HOX genes. Using hierarchical clustering analysis the HOX transcriptome was able to discriminate endothelial cells from 61 diverse human cell lines of various origins. In a separate publically available microarray dataset of 53 human endothelial cell lines, the HOX transcriptome additionally organized endothelial cells related to their organ or tissue of origin. Human tissue staining for HOXD8 and HOXD9 confirmed endothelial expression and also supported increased microvascular expression of these HOXs. Together these observations suggest a significant involvement of HOX genes in endothelial cell positional identity.
Collapse
Affiliation(s)
- Mark Toshner
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
- Papworth Hospital, Cambridge, United Kingdom
| | - Benjamin J. Dunmore
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Eoin F. McKinney
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Paola Caruso
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Paul D. Upton
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - John P. Waters
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Mark L. Ormiston
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jeremy N. Skepper
- Department of Physiology and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Gerard Nash
- School of Clinical and Experimental Medicine, Birmingham University, Birmingham, United Kingdom
| | - Amer A. Rana
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Toshner M, Dunmore BJ, McKinney EF, Southwood M, Caruso P, Upton PD, Waters JP, Ormiston ML, Skepper JN, Nash G, Rana AA, Morrell NW. Transcript analysis reveals a specific HOX signature associated with positional identity of human endothelial cells. PLoS One 2014; 1:430-5. [PMID: 23213434 PMCID: PMC3507213 DOI: 10.1242/bio.2012039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The distinct topographic Hox expression patterns observed in vascular smooth muscle cells (VSMCs) of the adult cardiovascular system suggest that these transcriptional regulators are critical for maintaining region-specific physiological properties of blood vessels. To test this proposition, we expanded the vascular Hoxc11 expression domain normally restricted to the lower limbs by utilizing an innovative integrated tetracycline regulatory system and Transgelin promoter elements to induce Hoxc11 expression universally in VSMCs of transgenic mice. Ectopic Hoxc11 expression in carotid arteries, aortic arch and descending aorta resulted in drastic vessel wall remodeling involving elastic laminae fragmentation, medial smooth muscle cell loss, and intimal lesion formation. None of these alterations were observed upon induction of Hoxc11 transgene expression in the femoral artery, i.e. the natural Hoxc11 activity domain, although this vessel was greatly enlarged, comparable to the topographically restricted vascular changes seen in Hoxc11−/− mice. To begin defining Hoxc11-controlled pathways of vascular remodeling, we performed immunolabeling studies in conjunction with co-transfection and chromatin immunoprecipitation (ChIP) assays using mouse vascular smooth muscle (MOVAS) cells. The results suggest direct transcriptional control of two members of the matrix metalloproteinase (Mmp) family, including Mmp2 and Mmp9 that are known as key players in the inception and progression of vascular remodeling events. In summary, the severe vascular abnormalities resulting from the induced dysregulated expression of a Hox gene with regional vascular patterning functions suggest that proper Hox function and regulation is critical for maintaining vascular functional integrity.
Collapse
Affiliation(s)
- Mark Toshner
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; Papworth Hospital, Cambridge, United Kingdom
| | - Benjamin J Dunmore
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Eoin F McKinney
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Paola Caruso
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Paul D Upton
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - John P Waters
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Mark L Ormiston
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jeremy N Skepper
- Department of Physiology and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Gerard Nash
- School of Clinical and Experimental Medicine, Birmingham University, Birmingham, United Kingdom
| | - Amer A Rana
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
27
|
Portalska KJ, Groen N, Krenning G, Georgi N, Mentink A, Harmsen MC, van Blitterswijk C, de Boer J. The effect of donor variation and senescence on endothelial differentiation of human mesenchymal stromal cells. Tissue Eng Part A 2013; 19:2318-29. [PMID: 23676150 DOI: 10.1089/ten.tea.2012.0646] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Application of autologous cells is considered for a broad range of regenerative therapies because it is not surrounded by the immunological and ethical issues of allo- or xenogenic cells. However, isolation, expansion, and application of autologous cells do suffer from variability in therapeutic efficacy due to donor to donor differences and due to prolonged culture. One important source of autologous cells is mesenchymal stromal cells (MSCs), which can differentiate toward endothelial-like cells, thus making them an ideal candidate as cell source for tissue vascularization. Here we screened MSCs from 20 donors for their endothelial differentiation capacity and correlated it with the gene expression profile of the whole genome in the undifferentiated state. Cells of all donors were able to form tubes on Matrigel and induced the expression of endothelial genes, although with quantitative differences. In addition, we analyzed the effect of prolonged in vitro expansion on the multipotency of human MSCs and found that endothelial differentiation is only mildly sensitive to expansion-induced loss of differentiation as compared to osteogenic and adipogenic differentiation. Our results show the robustness of the endothelial differentiation protocol and the gene expression data give insight in the differences in endothelial differentiation between donors.
Collapse
Affiliation(s)
- Karolina Janeczek Portalska
- 1 Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pathirage NA, Cocquebert M, Sadovsky Y, Abumaree M, Manuelpillai U, Borg A, Keogh RJ, Brennecke SP, Evain-Brion D, Fournier T, Kalionis B, Murthi P. Homeobox gene transforming growth factor β-induced factor-1 (TGIF-1) is a regulator of villous trophoblast differentiation and its expression is increased in human idiopathic fetal growth restriction. Mol Hum Reprod 2013; 19:665-75. [PMID: 23761267 DOI: 10.1093/molehr/gat042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abnormal trophoblast function is associated with human fetal growth restriction (FGR). Targeted disruption of homeobox gene transforming growth β-induced factor (TGIF-1) results in placental dysfunction in the mouse. The role of human TGIF-1 in placental cell function is unknown. The aims of this study were to determine the expression of TGIF-1 in human idiopathic FGR-affected placentae compared with gestation-matched controls (GMC), to elucidate the functional role of TGIF-1 in trophoblasts and to identify its downstream targets. Real-time PCR and immunoblotting revealed that TGIF-1 mRNA and protein expression was significantly increased in FGR-affected placentae compared with GMC (n = 25 in each group P < 0.05). Immunoreactive TGIF-1 was localized to the villous cytotrophoblasts, syncytiotrophoblast, microvascular endothelial cells and in scattered stromal cells in both FGR and GMC. TGIF-1 inactivation in BeWo cells using two independent siRNA resulted in significantly decreased mRNA and protein of trophoblast differentiation markers, human chorionic gonadotrophin (CGB/hCG), syncytin and 3β-hydroxysteroid dehydrogenase/3β-honest significant difference expression. Our data demonstrate that homeobox gene TGIF-1 is a potential up-stream regulator of trophoblast differentiation and the altered TGIF-1 expression may contribute to aberrant villous trophoblast differentiation in FGR.
Collapse
Affiliation(s)
- Niroshani A Pathirage
- Department of Perinatal Medicine Pregnancy Research Centre, and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Locked Bag 300, Parkville, VIC 3052, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Murthi P, Kalionis B, Rajaraman G, Keogh RJ, Da Silva Costa F. The role of homeobox genes in the development of placental insufficiency. Fetal Diagn Ther 2012; 32:225-30. [PMID: 22906990 DOI: 10.1159/000339657] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/21/2012] [Indexed: 11/19/2022]
Abstract
Intrauterine growth restriction (IUGR) is an adverse pregnancy outcome associated with significant perinatal and pediatric morbidity and mortality, and an increased risk of chronic disease later in adult life. While a number of maternal, fetal and environmental factors are known causes of IUGR, the majority of IUGR cases are of unknown cause. These IUGR cases are frequently associated with placental insufficiency, possibly as a result of placental maldevelopment. Understanding the molecular mechanisms of abnormal placental development in IUGR associated with placental insufficiency is therefore of increasing importance. Here, we review our understanding of transcriptional control of normal placental development as well as human IUGR associated with placental insufficiency. We also assess the potential for understanding transcriptional control as a means for revealing new molecular targets for the detection, diagnosis and clinical management of IUGR associated with placental insufficiency.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Perinatal Medicine, Pregnancy Research Centre, and Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Vic., Australia
| | | | | | | | | |
Collapse
|
30
|
Dunk C, Roggensack A, Cox B, Perkins J, Åsenius F, Keating S, Weksberg R, Kingdom J, Adamson S. A distinct microvascular endothelial gene expression profile in severe IUGR placentas. Placenta 2012; 33:285-93. [DOI: 10.1016/j.placenta.2011.12.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 12/22/2011] [Accepted: 12/27/2011] [Indexed: 01/13/2023]
|
31
|
Abstract
By its location between maternal and fetal bloodstreams the human placenta not only handles the materno-fetal transport of nutrients and gases, but may also be exposed to intrauterine conditions adversely affecting placental and fetal development. Such adverse conditions exist in pregnancies complicated by gestational diabetes mellitus (GDM), and have been associated with alterations in placental anatomy and physiology. These alterations are mainly based on changes on the micro-anatomical and/or even molecular level including aberrant villous vascularization, a disbalance of vasoactive molecules, and enhanced oxidative stress. The consequence thereof may be impaired fetal oxygenation and changes in transplacental nutrient supply. Although transplacental glucose flux is flow limited and independent of glucose transporter availability, transport of essential and nonessential amino acids and expression of genes involved in lipid transport and metabolism are significantly affected by GDM.
Collapse
Affiliation(s)
- M Gauster
- Institute of Cell Biology, Histology and Embryology, Center for Molecular Medicine, Medical University of Graz, Graz, Austria
| | | | | | | |
Collapse
|
32
|
Homeobox gene Distal-Less 3 is a regulator of villous cytotrophoblast differentiation and its expression is increased in human idiopathic foetal growth restriction. J Mol Med (Berl) 2011; 90:273-84. [DOI: 10.1007/s00109-011-0836-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/18/2011] [Accepted: 11/03/2011] [Indexed: 10/15/2022]
|
33
|
Wang Q, Zhu J, Zou L, Yang Y. Role of axonal guidance factor netrin-1 in human placental vascular growth. ACTA ACUST UNITED AC 2011; 31:246-250. [PMID: 21505994 DOI: 10.1007/s11596-011-0261-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Indexed: 11/28/2022]
Abstract
This study investigated the role of netrin-1 in placental vascular development. In vitro rat aortic ring assay and in vivo Matrigel plug assay were conducted to exmaine the effect of netrin-1 on angiogenesis. Human placental microvascular endothelial cells (HPMECs) were isolated and cultured and their viability, migration and tubular formation were studied, in order to examine the effects of netrin-1. The results showed that netrin-1 potently stimulated neovascularization in a mouse Matrigel plug in vivo and the sprouting of endothelial cells in rat aortic rings in vitro. In addition, netrin-1 enhanced the viability, migration and tube formation of HPMECs. Our study suggested that netrin-1 could significantly promote the formation of blood vessels of human placenta and may be a potential target for developing new therapeutic strategies for placental vasculature-related diseases.
Collapse
Affiliation(s)
- Qianhua Wang
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jianwen Zhu
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Zou
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yun Yang
- Department of Gynecology and Obstetrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
34
|
The role of placental homeobox genes in human fetal growth restriction. J Pregnancy 2011; 2011:548171. [PMID: 21547091 PMCID: PMC3087155 DOI: 10.1155/2011/548171] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 02/17/2011] [Indexed: 12/04/2022] Open
Abstract
Fetal growth restriction (FGR) is an adverse pregnancy outcome associated with significant perinatal and paediatric morbidity and mortality, and an increased risk of chronic disease later in adult life. One of the key causes of adverse pregnancy outcome is fetal growth restriction (FGR). While a number of maternal, fetal, and environmental factors are known causes of FGR, the majority of FGR cases remain idiopathic. These idiopathic FGR pregnancies are frequently associated with placental insufficiency, possibly as a result of placental maldevelopment. Understanding the molecular mechanisms of abnormal placental development in idiopathic FGR is, therefore, of increasing importance. Here, we review our understanding of transcriptional control of normal placental development and abnormal placental development associated with human idiopathic FGR. We also assess the potential for understanding transcriptional control as a means for revealing new molecular targets for the detection, diagnosis, and clinical management of idiopathic FGR.
Collapse
|
35
|
Sobrevia L, Abarzúa F, Nien JK, Salomón C, Westermeier F, Puebla C, Cifuentes F, Guzmán-Gutiérrez E, Leiva A, Casanello P. Review: Differential placental macrovascular and microvascular endothelial dysfunction in gestational diabetes. Placenta 2011; 32 Suppl 2:S159-64. [PMID: 21215450 DOI: 10.1016/j.placenta.2010.12.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 12/09/2010] [Accepted: 12/09/2010] [Indexed: 10/18/2022]
Abstract
Human endothelial dysfunction is a common feature in many diseases of pregnancy, such as gestational diabetes (GD). Metabolic changes include abnormal synthesis of nitric oxide (NO) and abnormal membrane transport of l-arginine and adenosine in primary cultures of human umbilical vein (HUVEC, macrovascular) and placental microvillus (hPMEC, microvascular) endothelial cells. These alterations are associated with modifications in the expression and activity of endothelial (eNOS) and inducible (iNOS) NO synthases, respectively, an effect that is maintained at least up to passage 5 in culture. HUVEC and hPMEC exhibit expression and activity of the human cationic amino acid transporter 1 (hCAT-1), equilibrative nucleoside transporters 1 (hENT1) and hENT2, as well as the corresponding SLC7A1, SLC29A1 and SLC29A2 gene promoter activities. Altered gene expression results from increased NO level, protein kinase C, mitogen-activated protein kinases, and hCHOP-C/EBPα transcription factor activation. Reduced ENT-mediated adenosine transport in GD is associated with stimulation of the l-arginine/NO pathway, and mainly due to reduced expression and activity of hENT1. In addition, hENT2 activity seems able to restore the reduced adenosine transport in GD. Additionally, insulin exerts a differential modulation of endothelial cells from macrocirculation compared with microcirculation, possibly due to expression of different insulin receptor isoforms. It is suggested that a common functional characteristic leading to changes in the bioavailability of adenosine and metabolism of l-arginine is evidenced by human fetal micro and macrovascular endothelium in GD.
Collapse
Affiliation(s)
- L Sobrevia
- Division of Obstetrics and Gynecology, Medical Research Centre (CIM), School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rajaraman G, Murthi P, Pathirage N, Brennecke SP, Kalionis B. Downstream targets of homeobox gene HLX show altered expression in human idiopathic fetal growth restriction. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:278-87. [PMID: 20008130 DOI: 10.2353/ajpath.2010.090187] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Fetal growth restriction (FGR), a clinically significant pregnancy disorder, is poorly understood at the molecular level. This study investigates idiopathic FGR associated with placental insufficiency. Previously, we showed that the homeobox gene HLX is expressed in placental trophoblast cells and that HLX expression is significantly decreased in human idiopathic FGR. Here, we used the novel approach of identifying downstream targets of HLX in cell culture to detect potentially important genes involved in idiopathic FGR. Downstream targets were revealed by decreasing HLX expression in cultured trophoblast cells with HLX-specific small interfering RNAs to model human idiopathic FGR and comparing these levels with controls using a real-time PCR-based gene profiling system. Changes in candidate HLX target mRNA levels were verified in an independent trophoblast cell line, and candidate target gene expression was assessed in human idiopathic FGR-affected placentae (n = 25) compared with gestation-matched controls (n = 25). The downstream targets RB1 and MYC, cell cycle regulatory genes, showed significantly increased mRNA levels in FGR-affected tissues compared with gestation-matched controls, whereas CCNB1, ELK1, JUN, and CDKN1 showed significantly decreased mRNA levels (n = 25, P < 0.001, t-test). The changes for RB1 and CDKN1C were verified by Western blot analysis in FGR-affected placentae compared with gestation-matched controls (n = 6). We conclude that cell cycle regulatory genes RB1, MYC, CCNB1, ELK1, JUN, and CDKN1C, which control important trophoblast cell functions, are targets of HLX.
Collapse
Affiliation(s)
- Gayathri Rajaraman
- Department of Obstetrics and Gynaecology, University of Melbourne, RWH Campus, 20 Flemington Rd., Parkville, Victoria 3052, Australia.
| | | | | | | | | |
Collapse
|
37
|
Yao Y, Shao ES, Jumabay M, Shahbazian A, Ji S, Boström KI. High-density lipoproteins affect endothelial BMP-signaling by modulating expression of the activin-like kinase receptor 1 and 2. Arterioscler Thromb Vasc Biol 2008; 28:2266-74. [PMID: 18948634 PMCID: PMC2709402 DOI: 10.1161/atvbaha.108.176958] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE High-density lipoproteins (HDL) have antiinflammatory effects on the vascular endothelium. Because bone morphogenetic proteins (BMPs) are known to be inflammatory mediators, we examined the effect of HDL on BMP signaling. METHODS AND RESULTS Increasing concentrations of HDL progressively enhanced expression of the activin-like kinase receptor (ALK)1 and ALK2 in human aortic endothelial cells as determined by real-time polymerase chain reaction and immunoblotting. Induction of ALK1 was a result of enhanced ALK2 expression as determined by siRNA interference, and was associated with increased levels of vascular endothelial growth factor (VEGF) and matrix Gla protein (MGP). The HDL-induction of ALK2 was dependent on BMP-signaling, and affected coregulation of the ALK2 gene by the homeodomain proteins MSX2, DLX3, and DLX5, as determined by reporter gene assays, siRNA interference, and chromatin immunoprecipitation. Apolipoprotein A-I transgenic mice, known to have high HDL and inhibition of atherogenesis, exhibited similar changes in aortic gene expression as seen in endothelial cells treated with HDL in vitro. CONCLUSIONS We conclude that HDL benefits the arterial wall by allowing for enhanced ALK1 and ALK2 signaling, resulting in an increase of VEGF and MGP, essential for endothelial cell survival and prevention of vascular calcification, respectively.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activin Receptors, Type II/genetics
- Activin Receptors, Type II/metabolism
- Animals
- Apolipoprotein A-I/genetics
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Base Sequence
- Bone Morphogenetic Protein 4/metabolism
- Bone Morphogenetic Protein 4/pharmacology
- Bone Morphogenetic Proteins/metabolism
- Calcium-Binding Proteins/metabolism
- Cattle
- Cells, Cultured
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Extracellular Matrix Proteins/metabolism
- Humans
- Lipoproteins, HDL/metabolism
- Lipoproteins, HDL/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Promoter Regions, Genetic
- RNA Interference
- RNA, Small Interfering/genetics
- Signal Transduction/drug effects
- Vascular Endothelial Growth Factor A/metabolism
- Matrix Gla Protein
Collapse
Affiliation(s)
- Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | | | | | | | | | | |
Collapse
|