1
|
Wong GP, Hartmann S, Nonn O, Cannon P, Nguyen TV, Kandel M, de Alwis N, Murphy CN, Pritchard N, Dechend R, Hannan NJ, Tong S, Simmons DG, Kaitu'u-Lino TJ. Stem Cell Markers LGR5, LGR4 and Their Immediate Signalling Partners are Dysregulated in Preeclampsia. Stem Cell Rev Rep 2024:10.1007/s12015-024-10831-2. [PMID: 39688759 DOI: 10.1007/s12015-024-10831-2] [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: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Leucine-rich repeat-containing G protein-coupled receptors 5/4 (LGR5/LGR4) are critical stem cell markers in epithelial tissues including intestine. They agonise wingless-related integration site (WNT) signalling. Until now, LGR5/LGR4 were uncharacterised in placenta, where analogous functions may exist. We characterised LGR5/LGR4, their ligands/targets in human placenta, with further assessments on dysregulation in preeclampsia/fetal growth restriction (FGR). LGR5 mRNA was unaltered in first trimester (n = 11), preterm (n = 9) and term (n = 11) placental lysate. LGR5 was enriched in human trophoblast stem cells (hTSCs) and downregulated with differentiation to extravillous trophoblasts (p < 0.0215) and syncytiotrophoblasts (p < 0.0350). In situ hybridisation localised LGR5 to unique, proliferative MKI67 + mononuclear trophoblasts underlying syncytium which concurred with proposed progenitor identities in single-cell transcriptomics. LGR5 expression was significantly reduced in placentas from early-onset preeclampsia (p < 0.0001, n = 81 versus n = 19 controls), late-onset preeclampsia (p = 0.0046, n = 20 versus n = 33 controls) and FGR (p = 0.0031, n = 34 versus n = 17 controls). LGR4 was elevated in first trimester versus preterm and term placentas (p = 0.0412), in placentas with early-onset preeclampsia (p = 0.0148) and in FGR (p = 0.0417). Transcriptomic analysis and in vitro hTSC differentiation to both trophoblast lineages suggested LGR4 increases with differentiation. Single-nucleus RNA sequencing of placental villous samples supported LGR5 and LGR4 localisation findings. Hypoxia/proinflammatory cytokine treatment modelling elements experienced by the placenta in placental insufficiency pathogenesis did not significantly alter LGR5/LGR4. Ligands R-spondins 1/3/4, and neutralising targets ring finger protein 43 (RNF43) and zinc and ring finger 3 (ZNRF3) were also reduced in placentas from preeclamptic pregnancies. This study is the first to describe LGR5/LGR4 and their signalling partner expression in human placenta. Their dysregulations in the preeclamptic placenta allude to disruptions to integral trophoblast stem cell function/differentiation that may occur during placental development related to WNT signalling.
Collapse
Affiliation(s)
- Georgia P Wong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| | - Sunhild Hartmann
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
| | - Olivia Nonn
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ping Cannon
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Manju Kandel
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ciara N Murphy
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ralf Dechend
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Klinikum, Berlin Buch, Germany
| | - Natalie J Hannan
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - David G Simmons
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
2
|
Nair S, Khambata K, Warke H, Bansal V, Patil A, Ansari Z, Balasinor NH. Methylation aberrations in partner spermatozoa and impaired expression of imprinted genes in the placentae of early-onset preeclampsia. Placenta 2024; 158:275-284. [PMID: 39527857 DOI: 10.1016/j.placenta.2024.10.068] [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: 06/25/2024] [Revised: 10/12/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Disturbed paternal epigenetic status of imprinted genes has been observed in infertility and recurrent spontaneous abortions. Shallow placentation has been associated with early-onset preeclampsia. Hence, the present study aimed to investigate the methylation patterns of imprinted genes involved in placental development, in the spermatozoa of partners of women experiencing preeclampsia. METHODS The study involved recruitment of couples into preeclampsia (n = 14) and control (n = 25) groups. Methylation analysis of imprinted gene differentially methylated regions (DMRs) and LINE1 repetitive element was carried out by pyrosequencing in the spermatozoa and placental villi. Global 5 mC levels in the spermatozoa were measured through ELISA. Expression of imprinted genes was quantified in the placental villi by real time qPCR. Association of birth weight with DNA methylation and gene expression was assessed. RESULTS KvDMR, PEG3 DMR, PEG10 DMR and DLK1-GTL2 IG-DMR were differentially methylated in the spermatozoa and placental villi of preeclampsia group. Global 5 mC content and LINE1 methylation levels did not differ between the spermatozoa of the two groups. Increased transcript levels of PEG3, IGF2, DLK1, PHLDA2 and CDKN1C were observed in the preeclamptic placental villi. Birth weight showed significant association with KvDMR, PEG10 DMR, DLK1-GTL2 IG-DMR and LINE1 methylation levels in the spermatozoa. DLK1 expression levels showed a negative association with birth weight. DISCUSSION The study highlighted the paternal contribution to early-onset preeclampsia, in the form of disrupted sperm DNA methylation patterns at imprinted gene loci. These loci, after further evaluation in future studies, could serve as sperm-based preeclampsia predictive markers, for couples planning pregnancy.
Collapse
Affiliation(s)
- Sweta Nair
- Department of Neuroendocrinology, National Institute for Research in Reproductive and Child Health, Mumbai, 400012, India
| | - Kushaan Khambata
- Department of Gamete Immunobiology, National Institute for Research in Reproductive and Child Health, Mumbai, 400012, India
| | - Himangi Warke
- Seth GS Medical College & KEM Hospital, Mumbai, 400012, India
| | - Vandana Bansal
- Nowrosjee Wadia Maternity Hospital, Mumbai, 400012, India
| | - Anushree Patil
- Department of Clinical Research, National Institute for Research in Reproductive and Child Health, Mumbai, 400012, India
| | - Zakiya Ansari
- Department of Neuroendocrinology, National Institute for Research in Reproductive and Child Health, Mumbai, 400012, India
| | - Nafisa H Balasinor
- Department of Neuroendocrinology, National Institute for Research in Reproductive and Child Health, Mumbai, 400012, India.
| |
Collapse
|
3
|
Pantho AF, Mohamed S, Govande JV, Rane R, Vora N, Kelso KR, Kuehl TJ, Lindheim SR, Uddin MN. Pravastatin Protects Cytotrophoblasts from Hyperglycemia-Induced Preeclampsia Phenotype. Cells 2024; 13:1534. [PMID: 39329718 PMCID: PMC11430553 DOI: 10.3390/cells13181534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
There are no effective therapies to prevent preeclampsia (PE). Pravastatin shows promise by attenuating processes associated with PE such as decreased cytotrophoblast (CTB) migration, aberrant angiogenesis, and increased oxidative stress. This study assesses the effects of pravastatin on hyperglycemia-induced CTB dysfunction. METHODS Human CTB cells were treated with 100, 150, 200, 300, or 400 mg/dL glucose for 48 h. Some cells were pretreated with pravastatin (1 µg/mL), while others were cotreated with pravastatin and glucose. The expression of urokinase plasminogen activator (uPA), plasminogen activator inhibitor 1 (PAI-1) mRNA, vascular endothelial growth factor (VEGF), placenta growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1), and soluble endoglin (sEng) were measured. CTB migration was assayed using a CytoSelect migration assay kit. Statistical comparisons were performed using an analysis of variance with Duncan's post hoc test. RESULTS The hyperglycemia-induced downregulation of uPA was attenuated in CTB cells pretreated with pravastatin at glucose levels > 200 mg/dL and cotreated at glucose levels > 300 mg/dL (p < 0.05). Hyperglycemia-induced decreases in VEGF and PlGF and increases in sEng and sFlt-1 were attenuated in both the pretreatment and cotreatment samples regardless of glucose dose (p < 0.05). Pravastatin attenuated hyperglycemia-induced dysfunction of CTB migration. CONCLUSIONS Pravastatin mitigates stress signaling responses in hyperglycemic conditions, weakening processes leading to abnormal CTB migration and invasion associated with PE in pregnancy.
Collapse
Affiliation(s)
- Ahmed F. Pantho
- Artemis Biotechnologies LLC, Temple, TX 76504, USA; (A.F.P.); (T.J.K.)
| | - Sara Mohamed
- Baylor Scott & White Health, Temple, TX 76508, USA; (S.M.); (N.V.); (K.R.K.); (S.R.L.)
| | | | - Riddhi Rane
- Texas A&M University College of Medicine, College Station, TX 77807, USA;
| | - Niraj Vora
- Baylor Scott & White Health, Temple, TX 76508, USA; (S.M.); (N.V.); (K.R.K.); (S.R.L.)
| | - Kelsey R. Kelso
- Baylor Scott & White Health, Temple, TX 76508, USA; (S.M.); (N.V.); (K.R.K.); (S.R.L.)
| | - Thomas J. Kuehl
- Artemis Biotechnologies LLC, Temple, TX 76504, USA; (A.F.P.); (T.J.K.)
| | - Steven R. Lindheim
- Baylor Scott & White Health, Temple, TX 76508, USA; (S.M.); (N.V.); (K.R.K.); (S.R.L.)
| | - Mohammad N. Uddin
- Artemis Biotechnologies LLC, Temple, TX 76504, USA; (A.F.P.); (T.J.K.)
- Baylor Scott & White Health, Temple, TX 76508, USA; (S.M.); (N.V.); (K.R.K.); (S.R.L.)
- Texas A&M University College of Medicine, College Station, TX 77807, USA;
| |
Collapse
|
4
|
Liu H, Yu L, Ding Y, Peng M, Deng Y. Progesterone Enhances the Invasion of Trophoblast Cells by Activating PI3K/AKT Signaling Pathway to Prevent Preeclampsia. Cell Transplant 2023; 32:9636897221145682. [PMID: 36593749 PMCID: PMC9830574 DOI: 10.1177/09636897221145682] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We aimed to explore whether the effect of progesterone on preeclampsia via the PI3K/AKT signaling pathway. First, we studied the role of progesterone in preeclampsia patients and HTR-8/Svneo cells by adding progesterone. Then PI3K inhibitor LY294002 was added. The effects of progesterone on preeclampsia were also studied in animals by constructing a preeclampsia rat model. CCK-8 and Transwell assay were applied to measure cell viability and invasion ability. ELISA was performed to measure progesterone, MMP-2, MMP-9, pro-inflammatory factors TNF-α, IL-1β, and anti-inflammatory factors IL-4, IL-10, and IL-13 levels. HE staining was used to detect the pathological changes in uterine spiral artery. Western blot was performed to detect Cyclin D1, PCNA, MMP-2, MMP-9, inflammatory factors TNF-α, IL-1β, IL-4, IL-10, IL-13, and PI3K/AKT signaling pathway related proteins AKT, p-AKT, PI3K, and p-PI3K expressions. Progesterone could reduce blood pressure and urine protein in pregnant women with preeclampsia. TNF-α and IL-1β levels were decreased, but IL-4, IL-10, IL-13, cyclin D1, and PCNA levels were increased in pregnant women with preeclampsia after using progesterone. After the use of progesterone, the symptoms of the PE model group were improved. Among them, the lumen of the placental uterine spiral artery was enlarged, and the fibrinoid necrosis of the uterine wall and acute atherosclerotic lesions were relieved. In addition, progesterone promoted HTR-8/Svneo cells proliferation and invasion. However, high expression of MMP-2, MMP-9, p-AKT, and p-PI3K in Normal and preeclampsia groups caused by progesterone was weakened after adding LY294002, indicating that progesterone could activate PI3K/AKT signaling pathway to regulate HTR-8/Svneo cells. Progesterone decreased urine protein and blood pressure of preeclampsia rats in a concentration-dependent manner. Moreover, progesterone activated the PI3K/AKT signaling pathway and inhibited the inflammatory response in preeclampsia rats.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ling Yu
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yiling Ding
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China,Yiling Ding, Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Renmin Middle Road 139, Changsha 410005, Hunan, China.
| | - Mei Peng
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yali Deng
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Functional Analysis of p21 Cip1/CDKN1A and Its Family Members in Trophoblastic Cells of the Placenta and Its Roles in Preeclampsia. Cells 2021; 10:cells10092214. [PMID: 34571867 PMCID: PMC8465116 DOI: 10.3390/cells10092214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/24/2022] Open
Abstract
Preeclampsia (PE), a gestational hypertensive disease originating from the placenta, is characterized by an imbalance of various cellular processes. The cell cycle regulator p21Cip1/CDKN1A (p21) and its family members p27 and p57 regulate signaling pathways fundamental to placental development. The aim of the present study was to enlighten the individual roles of these cell cycle regulators in placental development and their molecular involvement in the pathogenesis of PE. The expression and localization of p21, phospho-p21 (Thr-145), p27, and p57 was immunohistochemically analyzed in placental tissues from patients with early-onset PE, early-onset PE complicated by the HELLP (hemolysis, elevated liver enzymes and low platelet count) syndrome as well as late-onset PE compared to their corresponding control tissues from well-matched women undergoing caesarean sections. The gene level was evaluated using real-time quantitative PCR. We demonstrate that the delivery mode strongly influenced placental gene expression, especially for CDKN1A (p21) and CDKN1B (p27), which were significantly upregulated in response to labor. Cell cycle regulators were highly expressed in first trimester placentas and impacted by hypoxic conditions. In support of these observations, p21 protein was abundant in trophoblast organoids and hypoxia reduced its gene expression. Microarray analysis of the trophoblastic BeWo cell line depleted of p21 revealed various interesting candidate genes and signaling pathways for the fusion process. The level of p21 was reduced in fusing cytotrophoblasts in early-onset PE placentas and depletion of p21 led to reduced expression of fusion-related genes such as syncytin-2 and human chorionic gonadotropin (β-hCG), which adversely affected the fusion capability of trophoblastic cells. These data highlight that cell cycle regulators are important for the development of the placenta. Interfering with p21 influences multiple pathways related to the pathogenesis of PE.
Collapse
|
6
|
Molbay M, Özaydın-Goksu E, Kipmen-Korgun D, Unal A, Ozekinci M, Cebeci E, Maltepe E, Korgun ET. Human placental trophoblast progenitor cells (hTPCs) promote angiogenesis and neurogenesis after focal cerebral ischemia in rats. Int J Neurosci 2020; 132:258-268. [PMID: 32772609 DOI: 10.1080/00207454.2020.1807978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Reduction of blood flow below a threshold value in brain regions locally or globally is called cerebral ischemia and proper treatment requires either the restoration of normal blood flow and/or the administration of neuroprotective therapies. Human trophoblast progenitor cells (hTPCs) give rise to the placenta and are responsible for the invasion and vascular remodeling of the maternal vessels within the uterus. Here, we tested whether hTPCs promoted to differentiate along neural lineages may exhibit therapeutic properties in the setting of cerebral ischemia in vivo. MATERIALS AND METHODS Cerebral ischemia was generated in rats via middle cerebral artery occlusion and, after 24 h, hTPCs were injected systemically via tail vein. Animals were sacrified at Day 3 or 11. RESULTS TTC staining indicated that infarct volumes were smaller in hTPC treated animals. Visible myelin recovery was observed in the hTPC injected group with Luxol Fast Blue staining. On Day 11 after hTPC transplantation, DLX5 and VEGF expression, as well as 2 and 10 d after hTPC transplantation, NKX2.2 were significantly increased; while LHX6, Olig1, PDGFRα, VEGFR1 and VEGFR2 showed trends toward improved expression in brain tissue via immunoblot analysis. Neuron-like differentiated cells were positive for both NeuN and Cresyl Violet staining. CONCLUSION Here, we demonstrate for the first time that hTPCs enhance the expression of angiogenic and neurogenic factors in rat brain after stroke. Transplantation of hTPCs could form the basis of novel therapeutic approaches for the treatment of stroke in the clinical setting.
Collapse
Affiliation(s)
- Muge Molbay
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | | | - Dijle Kipmen-Korgun
- Department of Biochemistry, Akdeniz University School of Medicine, Antalya, Turkey
| | - Ali Unal
- Department of Neurology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Murat Ozekinci
- Department of Obstetrics and Gynecology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Erhan Cebeci
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Emin Turkay Korgun
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
7
|
Fantone S, Mazzucchelli R, Giannubilo SR, Ciavattini A, Marzioni D, Tossetta G. AT-rich interactive domain 1A protein expression in normal and pathological pregnancies complicated by preeclampsia. Histochem Cell Biol 2020; 154:339-346. [PMID: 32529396 DOI: 10.1007/s00418-020-01892-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
AT-rich interactive domain 1A (ARID1A, as known as BAF250a) is a subunit of human switch/sucrose nonfermentable chromatin remodeling complex with tumour suppressor function. Mutations of Arid1a have been reported in many human cancers and low expression of this protein has been correlated to a poor prognosis outcome in patients affected by some types of cancer. Although there are many studies regarding ARID1A functions in cancer, little is known about its role in regulating cell differentiation and normal tissues homeostasis. Here, we investigate ARID1A expression in normal placental tissues of first and third trimester of gestation and in pathological placental tissues of pregnancy complicated by preeclampsia (PE) and intrauterine growth restriction (IUGR) to evaluate a possible role of this protein in trophoblast differentiation. We found that ARID1A was specifically expressed in villous and extravillous cytotrophoblastic cells in normal placentas whereas syncytiotrophoblast was negative. Interestingly, ARID1A was expressed in both cytotrophoblastic cells and syncytiotrophoblast in placentas affected by PE and PE-IUGR. Moreover, ARID1A was also present in syncitial knots of pathological placentas. The present results indicate that ARID1A is a good marker of poor trophoblast differentiation in these pathologies, because the significant high positive staining in syncytiotrophoblast nuclei may suggest a poor differentiation of this trophoblast layer due to the cytotrophoblast cells fusion with the syncytiotrophoblast overlaying before arresting their cell cycle.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, United Hospitals, Ancona, Italy
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy
| |
Collapse
|
8
|
Farah O, Nguyen C, Tekkatte C, Parast MM. Trophoblast lineage-specific differentiation and associated alterations in preeclampsia and fetal growth restriction. Placenta 2020; 102:4-9. [PMID: 33218578 DOI: 10.1016/j.placenta.2020.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 12/26/2022]
Abstract
The human placenta is a poorly-understood organ, but one that is critical for proper development and growth of the fetus in-utero. The epithelial cell type that contributes to primary placental functions is called "trophoblast," including two main subtypes, villous and extravillous trophoblast. Cytotrophoblast and syncytiotrophoblast comprise the villous compartment and contribute to gas and nutrient exchange, while extravillous trophoblast invade and remodel the uterine wall and vessels, in order to supply maternal blood to the growing fetus. Abnormal differentiation of trophoblast contributes to placental dysfunction and is associated with complications of pregnancy, including preeclampsia (PE) and fetal growth restriction (FGR). This review describes what is known about the cellular organization of the placenta during both normal development and in the setting of PE/FGR. It also explains known trophoblast lineage-specific markers and pathways regulating their differentiation, and how these are altered in the setting of PE/FGR, focusing on studies which have used human placental tissues. Finally, it also highlights remaining questions and needed resources to advance this field.
Collapse
Affiliation(s)
- Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Calvin Nguyen
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chandana Tekkatte
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
9
|
Wen Z, Jiang R, Huang Y, Wen Z, Rui D, Liao X, Ling Z. Inhibition of lung cancer cells and Ras/Raf/MEK/ERK signal transduction by ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7). Respir Res 2019; 20:194. [PMID: 31443651 PMCID: PMC6708200 DOI: 10.1186/s12931-019-1165-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 08/13/2019] [Indexed: 12/11/2022] Open
Abstract
Background The aim of this study was to investigate the effects and mechanisms of ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7) on lung cancer cells. Methods The expression characteristics of ENTPD7 and its effect on the survival of lung cancer patients were analyzed by referring to The Cancer Genome Atlas (TCGA). Streptavidin-peroxidase (SP) staining was performed to detect the ENTPD7 protein in tumor tissues and adjacent tissues. Plasmid transfection technology was also applied to silence ENTPD7 gene. Crystal violet staining and flow cytometry were performed to determine cell proliferation and apoptosis. Tumor-bearing nude mice model was established to investigate the effect of sh-ENTPD7 on tumors. Results The results showed that patients with low levels of ENTPD7 had higher survival rates. ENTPD7 was up-regulated in lung cancer tissues and cells. Down-regulation of the expression of ENTPD7 inhibited proliferation but promoted apoptosis of lung cancer cell. Silencing ENTPD7 also inhibited the expression levels of Ras and Raf proteins and the phosphorylation of mitogen-activated protein kinase (MEK) and extracellular signal-regulated kinase (ERK). Tumor-bearing nude mice experiments showed that silencing ENTPD7 had an inhibitory effect on lung cancer cells. Conclusions ENTPD7 was overexpressed in lung cancer cells. Down-regulating ENTPD7 could inhibit lung cancer cell proliferation and promote apoptosis via inhibiting the Ras/Raf/MEK/ERK pathway.
Collapse
Affiliation(s)
- Zhongwei Wen
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Rongfang Jiang
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Ying Huang
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Zhineng Wen
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Dong Rui
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Xiaoxiao Liao
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Zhougui Ling
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China.
| |
Collapse
|
10
|
Li N, Meng G, Tong X, Sun F, Zeng J, Bai Y, Liang S, Hu H, Liu L, Han M, Lu C, Dai F. Effects of P27/Bmdacapo, in the CIP/KIP family, on cell proliferation, growth and development in the silkworm (Bombyx mori). Gene 2019; 700:31-37. [PMID: 30898712 DOI: 10.1016/j.gene.2019.02.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 10/27/2022]
Abstract
We investigated changes in expression of the CIP/KIP family-related genes and the cycle-dependent factors Pcna, Cdk4 and Cdk2 during the growth and development of mice, Drosophila and silkworms. When the organism was in a period of rapid development, the related genes of the CIP/KIP family had low expression level and the cell cycle-dependent genes were highly expressed. In mammals, the CIP/KIP family includes three genes, p21, p27/Dacapo and p57. However, only one gene, P27/Dacapo, exists in the CIP/KIP family in silkworm and the orthologous gene in the silkworm is named Bmdacapo. Down-regulation of Bmdacapo in silkworm embryos caused overdevelopment of the embryos and indicated that Bmdacapo can inhibit silkworm growth and development. Up-regulation of Bmdacapo in silkworm cells inhibited cell proliferation, whereas down-regulation of Bmdacapo promoted cell proliferation. In order to explore the mechanism of Bmdacapo regulated silkworm development and cell proliferation, the effect of Bmdacapo on cell cycle changes was examined. The results demonstrate that Bmdacapo was able to induce G1/S phase arrest in the cell cycle. In silkworm cells, Bmdacapo inhibits the expression of Pcna, CDK4 and CDK2, which affects the cell cycle and ultimately inhibits cell proliferation. This regulatory mechanism is particularly different from mammals.
Collapse
Affiliation(s)
- Niannian Li
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Gang Meng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Xiaoling Tong
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Fuze Sun
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Jie Zeng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Yanmin Bai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Shubo Liang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Hai Hu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Lanlan Liu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Minjin Han
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Cheng Lu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, College of Biotechnology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
11
|
Lv H, Zhou Q, Li L, Wang S. HLA-C promotes proliferation and cell cycle progression in trophoblast cells. J Matern Fetal Neonatal Med 2019; 34:512-518. [PMID: 31018729 DOI: 10.1080/14767058.2019.1611772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: The development of maternal-fetal immune tolerance and adequate trophoblast function are essential for the establishment and maintenance of pregnancy. Human leukocyte antigen (HLA), the major histocompatibility complex (MHC) antigen specific to humans, plays an important role in placentation and is involved in many pregnancy-associated diseases. HLA-C is the only classical MHC I gene expressed at the maternal-fetal interface. To investigate whether HLA-C plays an independent role in regulating the development of trophoblasts, we explored the effect of HLA-C expression on placental development.Methods: The role of HLA-C in the growth and migration of trophoblast JAR and HTR-8/Svneo cell lines was investigated after HLA-C-expressing lentivirus transfection.Results: The MTT assay and colony formation assay showed that HLA-C promoted cell proliferation. Furthermore, cell cycle analysis showed that HLA-C overexpression accelerated the transition of trophoblast cells from the G0/G1 phase to the S phase. However, FACS analysis and migration assay indicated that HLA-C had no significant influence on trophoblast apoptosis and migration.Conclusion: Our study demonstrated for the first time that besides being involved in immune tolerance, HLA-C can directly promote placental growth without interacting with immune cells, which could provide a new insight into studying the functions of HLA-C at the maternal-fetal interface.
Collapse
Affiliation(s)
- Hong Lv
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Qian Zhou
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lie Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shan Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
12
|
He GQ, Liu GY, Xu WM, Liao HJ, Liu XH, He GL. p57KIP2‑mediated inhibition of human trophoblast apoptosis and promotion of invasion in vitro. Int J Mol Med 2019; 44:281-290. [PMID: 31059007 DOI: 10.3892/ijmm.2019.4175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 04/19/2019] [Indexed: 11/06/2022] Open
Abstract
Placental hypoxia serves a role in the early stages of normal pregnancy and is involved in the pathophysiology of preeclampsia. Previously, it was suggested that p57kinase inhibitory protein (KIP)2 regulates the cell cycle during embryogenesis and apoptosis. Recent evidence has indicated that p57KIP2 is increased in preeclamptic placentas and absence of p57KIP2 induces preeclampsia‑type symptoms in rats. However, effects of p57KIP2 on apoptosis under hypoxic conditions remain to be elucidated. In the present study, HTR‑8/SVneo trophoblasts were cultured under hypoxic conditions (2% O2). Knockdown using small interfering (si)RNA and overexpression of p57KIP2 were utilized to explore the biological function of p57KIP2 in apoptosis and cell function in vitro. Furthermore, expression of p57KIP2 and apoptosis were evaluated by western blotting, flow cytometry and TUNEL assays, and the response of trophoblasts to hypoxia and the role of p57KIP2 in trophoblast migration and invasion was assessed. The role of p57KIP2 in the JNK signaling pathway in HTR‑8/SVneo trophoblasts was further studies. In vitro, protein expression of p57KIP2 was increased in HTR‑8/SVneo cells exposed to 2% O2. Exogenous p57KIP2 overexpression significantly decreased the expression of pro‑apoptosis proteins, including p53, Bax and cleaved caspase3, under hypoxic conditions for 24 h. In addition, knockdown of p57KIP2 increased the response to apoptosis following hypoxia for 24 h. The present study revealed that overexpression of p57KIP2 decreased the levels of phosphorylated‑JNK. JNK inhibitor treatment combined with the overexpression of p57KIP2 significantly decreased the levels of apoptosis and increased cell invasion and migration. Taken together, p57KIP2 knockdown significantly increased apoptosis in HTR‑8/SVneo cells exposed to 2% O2, whereas overexpression of p57KIP2 had opposite effects, mediated by the JNK/stress activated protein kinase (SAPK) signaling pathway. The results indicated that hypoxia‑induced expression of p57KIP2 promoted trophoblast migration and invasion by mediating the JNK/SAPK signaling pathway, which is crucial during placentation. These results may provide a novel molecular mechanism to understand the involvement of p57KIP2 in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Guo-Qian He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Guang-Yu Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wen-Ming Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hui-Juan Liao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xing-Hui Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Guo-Lin He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
13
|
Hong F, Zhao M, Zhang L, Feng L. Inhibition of Ezh2 In Vitro and the Decline of Ezh2 in Developing Midbrain Promote Dopaminergic Neurons Differentiation Through Modifying H3K27me3. Stem Cells Dev 2019; 28:649-658. [PMID: 30887911 DOI: 10.1089/scd.2018.0258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Epigenetic modifications play an important role in neural development. Trimethylated histone H3 at lysine 27 (H3K27me3) is a repressive epigenetic marker that mediates tissue development. In this study, we demonstrate that H3K27me3 and histone methyl transferase Ezh2 regulated the development of dopaminergic (DA) neurons in vitro and in vivo. We found that H3K27me3 increased during differentiation of ventral midbrain-derived neural stem cells (VM-NSCs). However, histone demethylase selective inhibitor GSK-J1 increased H3K27me3 level and decreased the expression of tyrosine hydroxylase. Treated with Ezh2-selective inhibitor EPZ005687 repressed the trimethylation of H3K27 and enhanced differentiation of DA neurons in VM-NSCs cultures. Furthermore, Ezh2 inhibition promoted the expression of DA neurons developmental-related factors by modifying H3K27 trimethylation on the relevant promoter regions. Moreover, the effect of Ezh2 inhibition-mediated DA neurons differentiation was blocked by the expression of shRNA specific for Nurr1. In vivo, Ezh2 decreased and resulted in a reduction of H3K27me3 in developing midbrain. Deletion of Ezh2 by RNA interference approach promoted differentiation of DA neurons during midbrain development. Overexpression of Ezh2 enhanced cell self-renewal and did not affect differentiation of DA neurons.
Collapse
Affiliation(s)
- Feng Hong
- 1 CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China.,2 Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Beijing, China
| | - Mengxue Zhao
- 1 CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China.,2 Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Beijing, China
| | - Lei Zhang
- 1 CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China
| | - Linyin Feng
- 1 CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China.,2 Department of Neuropharmacology, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Zhao WP, Wang HW, Liu J, Tan PP, Luo XL, Zhu SQ, Chen XL, Zhou BH. Positive PCNA and Ki-67 Expression in the Testis Correlates with Spermatogenesis Dysfunction in Fluoride-Treated Rats. Biol Trace Elem Res 2018; 186:489-497. [PMID: 29748930 DOI: 10.1007/s12011-018-1338-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 04/05/2018] [Indexed: 01/15/2023]
Abstract
The present study aimed to evaluate the effect of fluoride (F) on spermatogenesis in male rats. F- at 50 and 100 mg/L was administered for 70 days, after which the testicular and epididymis tissues were collected to observe the histopathological structure under a light microscope. The ultrastructure of the testis and sperm was also examined via transmission electron microscopy. The apoptosis of spermatogenic cells was measured through terminal deoxynucleotidyl transferase dUTP nick end labeling staining. The expression of proliferation factors, namely, proliferating cell nuclear antigen (PCNA) and Ki-67, in the testicular and epididymis tissues, were assayed through immunohistochemistry. F- at 50 and 100 mg/L significantly damaged the structure of the testis and epididymis, and the testis and sperm ultrastructure exhibited various changes, including mitochondrial swelling and vacuolization, and apsilated and raised sperm membrane. F treatment significantly increased spermatogenic cell apoptosis in the testis. PCNA (P < 0.01) and Ki-67 (P < 0.01) also presented positive expression in the testis. By comparison, no significant changes occurred in the epididymis. In summary, excessive F intake results in spermatogenesis dysfunction by damaging the testicular structure and inducing spermatogenic cell apoptosis in male rats. The positive expression level of PCNA and Ki-67 was a good response to spermatogenesis dysfunction.
Collapse
Affiliation(s)
- Wen-Peng Zhao
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Hong-Wei Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China.
| | - Jing Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Pan-Pan Tan
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Xiang-Long Luo
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Shi-Quan Zhu
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Xue-Li Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Bian-Hua Zhou
- College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China.
| |
Collapse
|
15
|
Kisanga EP, Tang Z, Guller S, Whirledge S. Glucocorticoid signaling regulates cell invasion and migration in the human first-trimester trophoblast cell line Sw.71. Am J Reprod Immunol 2018; 80:e12974. [DOI: 10.1111/aji.12974] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/12/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Edwina P. Kisanga
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale School of Medicine; New Haven CT USA
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale School of Medicine; New Haven CT USA
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale School of Medicine; New Haven CT USA
| | - Shannon Whirledge
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale School of Medicine; New Haven CT USA
| |
Collapse
|
16
|
Genetic and Epigenetic Control of CDKN1C Expression: Importance in Cell Commitment and Differentiation, Tissue Homeostasis and Human Diseases. Int J Mol Sci 2018; 19:ijms19041055. [PMID: 29614816 PMCID: PMC5979523 DOI: 10.3390/ijms19041055] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/31/2018] [Accepted: 03/31/2018] [Indexed: 12/28/2022] Open
Abstract
The CDKN1C gene encodes the p57Kip2 protein which has been identified as the third member of the CIP/Kip family, also including p27Kip1 and p21Cip1. In analogy with these proteins, p57Kip2 is able to bind tightly and inhibit cyclin/cyclin-dependent kinase complexes and, in turn, modulate cell division cycle progression. For a long time, the main function of p57Kip2 has been associated only to correct embryogenesis, since CDKN1C-ablated mice are not vital. Accordingly, it has been demonstrated that CDKN1C alterations cause three human hereditary syndromes, characterized by altered growth rate. Subsequently, the p57Kip2 role in several cell phenotypes has been clearly assessed as well as its down-regulation in human cancers. CDKN1C lies in a genetic locus, 11p15.5, characterized by a remarkable regional imprinting that results in the transcription of only the maternal allele. The control of CDKN1C transcription is also linked to additional mechanisms, including DNA methylation and specific histone methylation/acetylation. Finally, long non-coding RNAs and miRNAs appear to play important roles in controlling p57Kip2 levels. This review mostly represents an appraisal of the available data regarding the control of CDKN1C gene expression. In addition, the structure and function of p57Kip2 protein are briefly described and correlated to human physiology and diseases.
Collapse
|
17
|
HMG-box transcription factor 1: a positive regulator of the G1/S transition through the Cyclin-CDK-CDKI molecular network in nasopharyngeal carcinoma. Cell Death Dis 2018; 9:100. [PMID: 29367693 PMCID: PMC5833394 DOI: 10.1038/s41419-017-0175-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 11/09/2022]
Abstract
HMG-box transcription factor 1 (HBP1) has been reported to be a tumor suppressor in diverse malignant carcinomas. However, our findings provide a conclusion that HBP1 plays a novel role in facilitating nasopharyngeal carcinoma (NPC) growth. The Kaplan-Meier analysis indicates that high expression HBP1 and low miR-29c expression both are negatively correlated with the overall survival rates of NPC patients. HBP1 knockdown inhibits cellular proliferation and growth, and arrested cells in G1 phase rather than affected cell apoptosis via flow cytometry (FCM) analysis. Mechanistically, HBP1 induces the expression of CCND1 and CCND3 levels by binding to their promoters, and binds to CDK4, CDK6 and p16INK4A promoters while not affects their expression levels. CCND1 and CCND3 promote CCND1-CDK4, CCND3-CDK6, and CDK2-CCNE1 complex formation, thus, E2F-1 and DP-1 are activated to accelerate the G1/S transition in the cell cycle. MiR-29c is down-regulated and correlated with NPC tumorigenesis and progression. Luciferase assays confirms that miR-29c binds to the 3' untranslated region (3'-UTR) of HBP1. Introduction of pre-miR-29c decreased HBP1 mRNA and protein levels. Therefore, the high endogenous HBP1 expression might be attributed to the low levels of endogenous miR-29c in NPC. In addition, HBP1 knockdown and miR-29c agomir administration both decrease xenograft growth in nude mice in vivo. It is firstly reported that HBP1 knockdown inhibited the proliferation and metastasis of NPC, which indicates that HBP1 functions as a non-tumor suppressor gene in NPC. This study provides a novel potential target for the prevention of and therapies for NPC.
Collapse
|
18
|
Gu Y, Shi Y, Yang Q, Gu WW, He YP, Zheng HJ, Zhang X, Wang JM, Wang J. miR-3074-5p Promotes the Apoptosis but Inhibits the Invasiveness of Human Extravillous Trophoblast-Derived HTR8/SVneo Cells In Vitro. Reprod Sci 2017; 25:690-699. [PMID: 28826362 DOI: 10.1177/1933719117725823] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The objective of this study was to observe the effects of the overexpression of miR-3074-5p in human trophoblast cells in vitro. DESIGN Experimental in vitro study in HTR8/SVneo cells. METHODS HTR8/SVneo cells were transfected with miR-3074-5p mimic. The cell apoptosis and invasion were measured via flow cytometry and transwell assay, respectively. The expression levels of P53, Cyclin Dependent Kinase Inhibitor 1B (P27), BCL-2, BCL2 associated X (BAX), and BCL2 like 14 (BCL-G) in HTR8/SVneo cells were determined by Western blot. The alterations in gene expression profile of HTR8/SVneo cells were evaluated by complementary DNA microarray assay, and the differential expressions of dihydrolipoamide S-succinyltransferase (DLST), growth-associated protein 43 (GAP43), runt-related transcription factor 2 (RUNX2), and C-C type chemokine receptor 3 (CCR3) were validated by Western blot. Biofunctions of these differentially expressed genes were enriched by Gene Ontology analysis. RESULTS The overexpression of miR-3074-5p in HTR8/SVneo cells promoted cell apoptosis but inhibited cell invasion, being accompanied by the significantly elevated expressions of P27, BCL-2, and BCL-G. Meanwhile, an increased expression of P27 and P57 was also detected in a small sample size of placental villi of recurrent miscarriage (RM) patients. Totally, 411 genes and 397 genes were screened out, respectively, to be downregulated or upregulated at least by 2-folds in miR-3074-5p overexpressed HTR8/SVneo cells. These differentially expressed genes were involved in several important functions related to pregnancy. Subsequently, the reduced expressions of DLST and GAP43 proteins, as well as the increased expressions of CCR3 and RUNX2 proteins, were validated in miR-3074-5p overexpressed HTR8/SVneo cells. CONCLUSION These data suggested a potential contribution of miR-3074-5p in the pathogenesis of RM by disturbing the normal activities of trophoblast cells.
Collapse
Affiliation(s)
- Yan Gu
- 1 The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yan Shi
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China
| | - Qian Yang
- 3 School of Pharmacy, Fudan University, Shanghai, China
| | - Wen-Wen Gu
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China
| | - Ya-Ping He
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China
| | - Hua-Jun Zheng
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China
| | - Xuan Zhang
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China
| | - Jian-Mei Wang
- 1 The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jian Wang
- 2 Key Laboratory of Reproduction Regulation of NHFPC, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,3 School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Nomura Y, John RM, Janssen AB, Davey C, Finik J, Buthmann J, Glover V, Lambertini L. Neurodevelopmental consequences in offspring of mothers with preeclampsia during pregnancy: underlying biological mechanism via imprinting genes. Arch Gynecol Obstet 2017; 295:1319-1329. [PMID: 28382413 PMCID: PMC6058691 DOI: 10.1007/s00404-017-4347-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/07/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE Preeclampsia is known to be a leading cause of mortality and morbidity among mothers and their infants. Approximately 3-8% of all pregnancies in the US are complicated by preeclampsia and another 5-7% by hypertensive symptoms. However, less is known about its long-term influence on infant neurobehavioral development. The current review attempts to demonstrate new evidence for imprinting gene dysregulation caused by hypertension, which may explain the link between maternal preeclampsia and neurocognitive dysregulation in offspring. METHOD Pub Med and Web of Science databases were searched using the terms "preeclampsia," "gestational hypertension," "imprinting genes," "imprinting dysregulation," and "epigenetic modification," in order to review the evidence demonstrating associations between preeclampsia and suboptimal child neurodevelopment, and suggest dysregulation of placental genomic imprinting as a potential underlying mechanism. RESULTS The high mortality and morbidity among mothers and fetuses due to preeclampsia is well known, but there is little research on the long-term biological consequences of preeclampsia and resulting hypoxia on the fetal/child neurodevelopment. In the past decade, accumulating evidence from studies that transcend disciplinary boundaries have begun to show that imprinted genes expressed in the placenta might hold clues for a link between preeclampsia and impaired cognitive neurodevelopment. A sudden onset of maternal hypertension detected by the placenta may result in misguided biological programming of the fetus via changes in the epigenome, resulting in suboptimal infant development. CONCLUSION Furthering our understanding of the molecular and cellular mechanisms through which neurodevelopmental trajectories of the fetus/infant are affected by preeclampsia and hypertension will represent an important first step toward preventing adverse neurodevelopment in infants.
Collapse
Affiliation(s)
- Yoko Nomura
- Department of Psychology, Queens College, the City University of New York, 65-30 Kissena Blvd, Flushing, NY, 11367, USA.
- Graduate Center, the City University of New York, Flushing, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Rosalind M John
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | | - Charles Davey
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jackie Finik
- Department of Psychology, Queens College, the City University of New York, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jessica Buthmann
- Department of Psychology, Queens College, the City University of New York, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- Graduate Center, the City University of New York, Flushing, USA
| | | | - Luca Lambertini
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
20
|
Baker BC, Mackie FL, Lean SC, Greenwood SL, Heazell AEP, Forbes K, Jones RL. Placental dysfunction is associated with altered microRNA expression in pregnant women with low folate status. Mol Nutr Food Res 2017; 61. [PMID: 28105727 PMCID: PMC5573923 DOI: 10.1002/mnfr.201600646] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/27/2016] [Accepted: 01/10/2017] [Indexed: 12/21/2022]
Abstract
SCOPE Low maternal folate status during pregnancy increases the risk of delivering small for gestational age (SGA) infants, but the mechanistic link between maternal folate status, SGA, and placental dysfunction is unknown. microRNAs (miRNAs) are altered in pregnancy pathologies and by folate in other systems. We hypothesized that low maternal folate status causes placental dysfunction, mediated by altered miRNA expression. METHODS AND RESULTS A prospective observational study recruited pregnant adolescents and assessed third trimester folate status and placental function. miRNA array, QPCR, and bioinformatics identified placental miRNAs and target genes. Low maternal folate status is associated with higher incidence of SGA infants (28% versus 13%, p < 0.05) and placental dysfunction, including elevated trophoblast proliferation and apoptosis (p < 0.001), reduced amino acid transport (p < 0.01), and altered placental hormones (pregnancy-associated plasma protein A, progesterone, and human placental lactogen). miR-222-3p, miR-141-3p, and miR-34b-5p were upregulated by low folate status (p < 0.05). Bioinformatics predicted a gene network regulating cell turnover. Quantitative PCR demonstrated that key genes in this network (zinc finger E-box binding homeobox 2, v-myc myelocytomatosis viral oncogene homolog (avian), and cyclin-dependent kinase 6) were reduced (p < 0.05) in placentas with low maternal folate status. CONCLUSION This study supports that placental dysfunction contributes to impaired fetal growth in women with low folate status and suggests altered placental expression of folate-sensitive miRNAs and target genes as a mechanistic link.
Collapse
Affiliation(s)
- Bernadette C Baker
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Fiona L Mackie
- Centre of Women's and Newborn's Health & Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Samantha C Lean
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | | | - Karen Forbes
- Division of Reproduction and Early Development, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Wu Y, Fu X, Wang L. Identification of novel biomarkers for preeclampsia on the basis of differential expression network analysis. Exp Ther Med 2016; 12:201-207. [PMID: 27347039 PMCID: PMC4906647 DOI: 10.3892/etm.2016.3261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 02/11/2016] [Indexed: 12/27/2022] Open
Abstract
Preeclampsia (PE) is a severe pregnancy complication, which is a leading cause of maternal and fetal mortality. The present study aimed to screen potential biomarkers for the diagnosis and prediction of PE and to investigate the underlying mechanisms of PE development based on the differential expression network (DEN). The microarray datasets E-GEOD-6573 and E-GEOD-48424 were downloaded from the European Bioinformatics Institute database. Differentially expressed genes (DEGs) between the PE and normal groups were screened by Significant Analysis of Microarrays with the cutoff value of a |log2 fold change| of >2, and a false discovery rate of <0.05. The DEN was constructed based on the differential and non-differential interactions observed. In addition, genes with higher connectivity degrees in the DEN were identified on the basis of centrality analysis, while disease genes were also extracted from the DEN. In order to understand the functional roles of genes in DEN, Gene Ontology (GO) and pathway enrichment analyses were performed. The present results indicated that a total of 225 genes were considered as DEGs in the PE group, while 466 nodes and 314 gene interactions were involved in the DEN. Among these 466 nodes, 4 nodes with higher degrees were identified, including ubiquitin C (UBC), small ubiquitin-like modifier 1 (SUMO1), SUMO2 and RAD21 homolog (S. pombe) (RAD21). Notably, UBC was also found to be a disease gene. UBC, RAD21, SUMO2 and SUMO1 were markedly enriched in the regulation of programmed cell death, as well as in the regulation of apoptosis, cell cycle and chromosomal part. In conclusion, based on these results, we suggest that UBC, RAD21, SUMO2 and SUMO1 may be reliable biomarkers for the prediction of the development and progression of PE.
Collapse
Affiliation(s)
- Yufang Wu
- Department of Gynecology, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Xiuhua Fu
- Department of Obstetrics, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Lin Wang
- Department of Obstetrics and Gynecology, Xijing Hospital, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shanxi 710032, P.R. China
| |
Collapse
|
22
|
Liu M, Li M, Liu J, Wang H, Zhong D, Zhou H, Yang B. Elevated urinary urea by high-protein diet could be one of the inducements of bladder disorders. J Transl Med 2016; 14:53. [PMID: 26879937 PMCID: PMC4755000 DOI: 10.1186/s12967-016-0809-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/04/2016] [Indexed: 12/22/2022] Open
Abstract
Background Previous work found that urea accumulation in urothelial cells caused by urea transporter B knockout led to DNA damage and apoptosis that contributed to the carcinogenesis. The purpose of this study is to explore the potential connection between high urinary urea concentration and the bladder disorders. Methods A high protein diet rat model was conducted by feeding with 40 % protein diet. In-silico modeling and algorithm, based on the results of microarray and proteomics from the bladder urothelium, were used for the reconstruction of accurate cellular networks and the identification of novel master regulators in the high-protein diet rat model. Pathway and biological process enrichment analysis were used to characterize predicted targets of candidate mRNAs/proteins. The expression pattern of the most significant master regulators was evaluated by qPCR and immunohistochemistry. Results Based on the analysis of different expressed mRNAs/proteins, 15 significant ones (CRP, MCPT2, MCPT9, EPXH2, SERPING1, SRGN, CDKN1C, CDK6, CCNB1, PCNA, BAX, MAGEB16, SERPINE1, HSPA2, FOS) were highly identified and verified by qPCR and immunohistochemistry. They were involved in immune and inflammatory response, cell cycle arrest, apoptosis and pathways in cancer. These abnormally activated processes caused the bladder interstitial congestion and inflammatory infiltrates under the thinner urothelium, cell desquamation, cytoplasm vacuolization, nucleus swelling and malformation in the high-protein diet group. Conclusions We provided evidences that high urinary urea concentration caused by high-protein diet might be a potential carcinogenic factor in bladder.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| | - Jiangfeng Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| | - Hongkai Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China. .,Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.
| | - Dandan Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Lu, Haidian District, Beijing, 100191, China.
| |
Collapse
|
23
|
Effects of induced pluripotent stem cells-derived conditioned medium on the proliferation and anti-apoptosis of human adipose-derived stem cells. Mol Cell Biochem 2016; 413:69-85. [DOI: 10.1007/s11010-015-2640-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/23/2015] [Indexed: 01/09/2023]
|
24
|
Kaitu'u-Lino T, Hastie R, Cannon P, Nguyen H, Lee S, Hannan N, Tong S. Transcription factors E2F1 and E2F3 are expressed in placenta but do not regulate MMP14. Placenta 2015; 36:932-7. [DOI: 10.1016/j.placenta.2015.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 06/09/2015] [Accepted: 06/14/2015] [Indexed: 01/22/2023]
|
25
|
Janssen AB, Tunster SJ, Savory N, Holmes A, Beasley J, Parveen SAR, Penketh RJA, John RM. Placental expression of imprinted genes varies with sampling site and mode of delivery. Placenta 2015; 36:790-5. [PMID: 26162698 PMCID: PMC4535278 DOI: 10.1016/j.placenta.2015.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED Imprinted genes, which are monoallelically expressed by virtue of an epigenetic process initiated in the germline, are known to play key roles in regulating fetal growth and placental development. Numerous studies are investigating the expression of these imprinted genes in the human placenta in relation to common complications of pregnancy such as fetal growth restriction and preeclampsia. This study aimed to determine whether placental sampling protocols or other factors such as fetal sex, gestational age and mode of delivery may influence the expression of imprinted genes predicted to regulate placental signalling. METHODS Term placentas were collected from Caucasian women delivering at University Hospital of Wales or Royal Gwent Hospital within two hours of delivery. Expression of the imprinted genes PHLDA2, CDKN1C, PEG3 and PEG10 was assayed by quantitative real time PCR. Intraplacental gene expression was analysed (N = 5). Placental gene expression was compared between male (N = 11) and female (N = 11) infants, early term (N = 8) and late term (N = 10) deliveries and between labouring (N = 13) and non-labouring (N = 21) participants. RESULTS The paternally expressed imprinted genes PEG3 and PEG10 were resilient to differences in sampling site, fetal sex, term gestational age and mode of delivery. The maternally expressed imprinted gene CDKN1C was elevated over 2-fold (p < 0.001) in placenta from labouring deliveries compared with elective caesarean sections. In addition, the maternally expressed imprinted gene PHLDA2 was elevated by 1.8 fold (p = 0.01) in samples taken at the distal edge of the placenta compared to the cord insertion site. CONCLUSION These findings support the reinterpretation of existing data sets on these genes in relation to complications of pregnancy and further reinforce the importance of optimising and unifying placental collection protocols for future studies.
Collapse
Affiliation(s)
- A B Janssen
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK
| | - S J Tunster
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK
| | - N Savory
- Department of Obstetrics and Gynaecology, University Hospital Wales, Cardiff, Wales CF144XW, UK
| | - A Holmes
- Department of Obstetrics and Gynaecology, University Hospital Wales, Cardiff, Wales CF144XW, UK
| | - J Beasley
- Department of Obstetrics and Gynaecology, Royal Gwent Hospital, Newport, Wales NP202UB, UK
| | - S A R Parveen
- Department of Obstetrics and Gynaecology, Royal Gwent Hospital, Newport, Wales NP202UB, UK
| | - R J A Penketh
- Department of Obstetrics and Gynaecology, University Hospital Wales, Cardiff, Wales CF144XW, UK
| | - R M John
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK.
| |
Collapse
|
26
|
Guo X, Li S, Ji Q, Lian R, Chen J. Enhanced viability and neural differential potential in poor post-thaw hADSCs by agarose multi-well dishes and spheroid culture. Hum Cell 2015; 28:175-89. [PMID: 26054839 DOI: 10.1007/s13577-015-0116-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/09/2015] [Indexed: 01/06/2023]
Abstract
Human adipose-derived stem cells (hADSCs) are potential adult stem cells source for cell therapy. But hADSCs with multi-passage or cryopreservation often revealed poor growth performance. The aim of our work was to improve the activity of poor post-thaw hADSCs by simple and effective means. We describe here a simple method based on commercially available silicone micro-wells for creating hADSCs spheroids to improve viability and neural differentiation potential on poor post-thaw hADSCs. The isolated hADSCs positively expresse d CD29, CD44, CD105, and negatively expressed CD34, CD45, HLA-DR by flow cytometry. Meanwhile, they had adipogenic and osteogenic differentiation capacity. The post-thaw and post-spheroid hADSCs from poor growth status hADSCs showed a marked increase in cell proliferation by CKK-8 analysis, cell cycle analysis and Ki67/P27 quantitative polymerase chain reaction (qPCR) analysis. They also displayed an increase viability of anti-apoptosis by annexin v and propidium iodide assays and mitochondrial membrane potential assays. After 3 days of neural induction, the neural differentiation potential of post-thaw and post-spheroid hADSCs could be enhanced by qPCR analysis and western blotting analysis. These results suggested that the spheroid formation could improve the viability and neural differentiation potential of bad growth status hADSCs, which is conducive to ADSCs research and cell therapy.
Collapse
Affiliation(s)
- Xiaoling Guo
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Shanyi Li
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Qingshan Ji
- Department of Ophthalmology, Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, China
| | - Ruiling Lian
- Department of Ophthalmology, The First Clinical Medical College of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China. .,Eye Institute, Medical College of Jinan University, Jinan University, Guangzhou, China. .,Department of Ophthalmology, The First Clinical Medical College of Jinan University, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
27
|
Gu Y, He Y, Zhang X, Shi Y, Yang Q, Yu L, Sun Z, Zhang H, Wang J, Gao X, Wang J. Deficiency of monoclonal non-specific suppressor factor beta (MNSFB) promotes pregnancy loss in mice. Mol Reprod Dev 2015; 82:475-88. [DOI: 10.1002/mrd.22495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/21/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Yan Gu
- Shanghai Medical School; Fudan University; Shanghai China
| | - Yaping He
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| | - Xuan Zhang
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| | - Yan Shi
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| | - Qian Yang
- Shanghai Medical School; Fudan University; Shanghai China
| | - Lin Yu
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| | - Zhaogui Sun
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| | - Huiqing Zhang
- Shanghai Medical School; Fudan University; Shanghai China
| | - Jianmei Wang
- The Second Hospital of Tianjin Medical University; Tianjin China
| | - Xiang Gao
- MOE Key Laboratory Model Animal for Disease Study; Model Animal Research Center; Nanjing University; Nanjing Jiangsu China
| | - Jian Wang
- Shanghai Institute of Planned Parenthood Research; Shanghai China
| |
Collapse
|
28
|
Immunohistochemical protein expression profiling of growth- and apoptotic-related factors in relation to umbilical cord length. Early Hum Dev 2015; 91:291-7. [PMID: 25804127 DOI: 10.1016/j.earlhumdev.2015.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Umbilical cord (UC) alterations are related to fetal and neonatal deaths and late neurological complications. Abnormal UC length has been recognized as the most significant abnormality linked to unfavorable outcomes. Despite its importance, causal factors resulting in abnormally long or short UCs have yet to be established. The factors that govern UC length are largely unknown. Furthermore, there is a paucity of studies that assess molecular processes involved in the establishment of UC length. We hypothesize that UC length abnormalities in UC length are associated with altered protein expression patterns of known cell growth and/or apoptosis regulators. In this study we analyze diverse protein expression patterns in different UC cell types found in UCs of normal and abnormal length. METHODS An analytical observational study was carried out on fetal autopsies; diagnosed abnormal length UCs were compared to normal controls by gestational age. Immunohistochemical analysis of expression levels of growth and pro- and anti-apoptotic factors was performed. RESULTS We performed immunohistochemistry antibody tests against FAS, BAX, Ki67, cMyc, FGF2, TGFBR3, VEGF, Bcl2, p57 and IGF2 and analyzed UC cell expression patterns. We found significant differences in specific long and/or short cord cell types in comparison to those in normal cords. DISCUSSION Factors that determine UC length are still largely unknown; however, this study demonstrates significant specific cell type differences in protein expression patterns of several genes related to cell proliferation. This preliminary study provides strong supporting data to continue the search for molecular factors that determine UC length.
Collapse
|
29
|
Kaya B, Nayki U, Nayki C, Ulug P, Oner G, Gultekin E, Yildirim Y. Proliferation of trophoblasts and Ki67 expression in preeclampsia. Arch Gynecol Obstet 2014; 291:1041-6. [PMID: 25384521 DOI: 10.1007/s00404-014-3538-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 11/04/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND/AIMS Preeclampsia is a pregnancy-specific disease with the increased risk of maternal morbidity and mortality. It is characterised by placental vascular dysfunction. Despite the numerous studies on preeclampsia, studies evaluating proliferation of villous trophoblasts in preeclamptic placentas are limited. Ki67 is a proliferation marker that expresses in the nuclei of proliferating cells. In this study, we examined the proliferation of villous trophoblasts in placentas of preeclamptic patients by using Ki67 and compared it with placentas of normal pregnant patients. MATERIAL AND METHODS The current study is a prospective one, including 15 placentas from preeclamptic patients and 14 placentas from normal pregnancies as controls. For detection of proliferation in villous trophoblasts, Ki67 was used. RESULTS The Ki67 index was 11.48±1.67% in normal patients and 15.53±2.28% in preeclamptic patients. There was a difference in Ki67 index between the two groups (p < 0.001). CONCLUSION Our results support the opinion that trophoblasts undergo regeneration hyperplasia as a result of injuries arising on the villous surface in preeclampsia. Proliferation of trophoblasts may contribute the development of preeclampsia.
Collapse
Affiliation(s)
- Barış Kaya
- Department of Obstetrics and Gynaecology, Near East University, North Cyprus, Cyprus
| | | | | | | | | | | | | |
Collapse
|
30
|
Can M, Guven B, Bektas S, Arikan I. Oxidative stress and apoptosis in preeclampsia. Tissue Cell 2014; 46:477-81. [PMID: 25200618 DOI: 10.1016/j.tice.2014.08.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/07/2014] [Accepted: 08/18/2014] [Indexed: 12/30/2022]
Abstract
We aimed to determine the oxidative stress and antioxidant status in preeclamptic placenta. Also, we investigated the apoptotic index of villous trophoblast and proliferation index of cytotrophoblasts. The study included 32 pregnant with preeclampsia and 31 normotensive healthy pregnant women. Malondialdehyde (MDA) and total antioxidant status (TAS) levels were measured in the placenta. For detection of apoptosis and proliferation in trophoblast, apoptosis protease activating factor 1 (APAF-1) and Ki-67 were used. Placental MDA levels in preeclamptic women were significantly higher than normal pregnancies (p=0.002). There was no significant difference between the groups in the TAS levels of placenta (p=0.773). Also, the apoptotic index in villous trophoblasts increased (p<0.001), but proliferation index did not change in preeclampsia (p=0.850). Increased oxidative stress and apoptosis in pathological placenta are not balanced by antioxidant systems and proliferation mechanisms.
Collapse
Affiliation(s)
- Murat Can
- Bulent Ecevit University, Faculty of Medicine, Department of Biochemistry, Zonguldak, Turkey
| | - Berrak Guven
- Bulent Ecevit University, Faculty of Medicine, Department of Biochemistry, Zonguldak, Turkey.
| | - Sibel Bektas
- Bulent Ecevit University, Faculty of Medicine, Department of Pathology, Zonguldak, Turkey
| | - Ilker Arikan
- Bulent Ecevit University, Faculty of Medicine, Department of Gynaecology and Obstetrics, Zonguldak, Turkey
| |
Collapse
|