1
|
Long J, Huang Y, Wang G, Tang Z, Shan Y, Shen S, Ni X. Mitochondrial ROS Accumulation Contributes to Maternal Hypertension and Impaired Remodeling of Spiral Artery but Not IUGR in a Rat PE Model Caused by Maternal Glucocorticoid Exposure. Antioxidants (Basel) 2023; 12:antiox12050987. [PMID: 37237853 DOI: 10.3390/antiox12050987] [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] [Received: 02/09/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Increased maternal glucocorticoid levels have been implicated as a risk factor for preeclampsia (PE) development. We found that pregnant rats exposed to dexamethasone (DEX) showed hallmarks of PE features, impaired spiral artery (SA) remodeling, and elevated circulatory levels of sFlt1, sEng IL-1β, and TNFα. Abnormal mitochondrial morphology and mitochondrial dysfunction in placentas occurred in DEX rats. Omics showed that a large spectrum of placental signaling pathways, including oxidative phosphorylation (OXPHOS), energy metabolism, inflammation, and insulin-like growth factor (IGF) system were affected in DEX rats. MitoTEMPO, a mitochondria-targeted antioxidant, alleviated maternal hypertension and renal damage, and improved SA remodeling, uteroplacental blood flow, and the placental vasculature network. It reversed several pathways, including OXPHOS and glutathione pathways. Moreover, DEX-induced impaired functions of human extravillous trophoblasts were associated with excess ROS caused by mitochondrial dysfunction. However, scavenging excess ROS did not improve intrauterine growth retardation (IUGR), and elevated circulatory sFlt1, sEng, IL-1β, and TNFα levels in DEX rats. Our data indicate that excess mitochondrial ROS contributes to trophoblast dysfunction, impaired SA remodeling, reduced uteroplacental blood flow, and maternal hypertension in the DEX-induced PE model, while increased sFlt1 and sEng levels and IUGR might be associated with inflammation and an impaired energy metabolism and IGF system.
Collapse
Affiliation(s)
- Jing Long
- Department of Gynecology and Obstetrics, Xiangya Hospital Central South University, Changsha 410008, China
- National International Joint Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha 410008, China
| | - Yan Huang
- Reproductive Medicine Center, General Hospital of Southern Theatre Command, Guangzhou 510010, China
| | - Gang Wang
- Department of Physiology, Naval Medical University, Shanghai 200433, China
| | - Zhengshan Tang
- National International Joint Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha 410008, China
| | - Yali Shan
- Department of Gynecology and Obstetrics, Xiangya Hospital Central South University, Changsha 410008, China
- National International Joint Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha 410008, China
| | - Shiping Shen
- Department of Gynecology and Obstetrics, Xiangya Hospital Central South University, Changsha 410008, China
- National International Joint Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha 410008, China
| | - Xin Ni
- National International Joint Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha 410008, China
| |
Collapse
|
2
|
Mei S, Li L, Zhou X, Xue C, Livingston MJ, Wei Q, Dai B, Mao Z, Mei C, Dong Z. Susceptibility of renal fibrosis in diabetes: Role of hypoxia inducible factor-1. FASEB J 2022; 36:e22477. [PMID: 35881071 PMCID: PMC9386694 DOI: 10.1096/fj.202200845r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 01/09/2023]
Abstract
Diabetes may prevent kidney repair and sensitize the kidney to fibrosis or scar formation. To test this possibility, we examined renal fibrosis induced by unilateral ureteral obstruction (UUO) in diabetic mouse models. Indeed, UUO induced significantly more renal fibrosis in both Akita and STZ-induced diabetic mice than in nondiabetic mice. The diabetic mice also had more apoptosis and interstitial macrophage infiltration during UUO. In vitro, hypoxia induced higher expression of the fibrosis marker protein fibronectin in high glucose-conditioned renal tubular cells than in normal glucose cells. Mechanistically, hypoxia induced significantly more hypoxia-inducible factor-1 α (HIF-1 α) in high glucose cells than in normal glucose cells. Inhibition of HIF-1 attenuated the expression of fibronectin induced by hypoxia in high-glucose cells. Consistently, UUO induced significantly higher HIF-1α expression along with fibrosis in diabetic mice kidneys than in nondiabetic kidneys. The increased expression of fibrosis induced by UUO in diabetic mice was diminished in proximal tubule-HIF-1α-knockout mice. Together, these results indicate that diabetes sensitizes kidney tissues and cells to fibrogenesis probably by enhancing HIF-1 activation.
Collapse
Affiliation(s)
- Shuqin Mei
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Lin Li
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Cheng Xue
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Bing Dai
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhiguo Mao
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Changlin Mei
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| |
Collapse
|
3
|
Tong S, Kaitu’u-Lino TJ, Hastie R, Brownfoot F, Cluver C, Hannan N. Pravastatin, proton-pump inhibitors, metformin, micronutrients, and biologics: new horizons for the prevention or treatment of preeclampsia. Am J Obstet Gynecol 2022; 226:S1157-S1170. [PMID: 32946849 DOI: 10.1016/j.ajog.2020.09.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
There has been increasing research momentum to identify new therapeutic agents for the prevention or treatment of preeclampsia, drugs that can affect the underlying disease pathophysiology. Molecular targets of candidate treatments include oxidative stress, antiangiogenic factors, and the angiotensin, nitric oxide, and proinflammatory pathways. The proposed treatments undergoing preclinical and clinical trial evaluation are thought to act on placental or endothelial disease or both. Most have adopted the pragmatic strategy of repurposing drugs. Of all the therapeutic agents proposed, pravastatin has received the most interest. There are preclinical studies showing that it has pleiotropic actions that favorably impact on multiple molecular targets and can resolve a preeclampsia phenotype in many animal models. An early phase clinical trial suggests that it may have therapeutic activity. Several large prevention trials are planned or ongoing and, when completed, could definitively address whether pravastatin can prevent preeclampsia. Proton-pump inhibitors, metformin, and sulfasalazine are other drugs with preclinical evidence of multiple molecular actions that could resolve the pathophysiology of preeclampsia. These agents are also currently being evaluated in clinical trials. There have been many recent preclinical studies identifying the potential of numerous natural compounds to treat preeclampsia, such as plant extracts and micronutrients that have potent anti-inflammatory or antioxidant activity. Recent preclinical studies have also proposed novel molecular-targeted strategies, such as monoclonal antibodies targeting tumor necrosis factor alpha, placental growth factor, and short interfering RNA technology, to silence the gene expression of soluble fms-like tyrosine kinase-1 or angiotensinogen. Other treatment approaches that have transitioned to human trials (ranging from single-arm to phase III trials that have been completed or are ongoing) include folic acid, nitric oxide donors (such as L-arginine), recombinant antithrombin III, digoxin immune antigen-binding fragment, and melatonin. There have been case series showing the removal of circulating soluble fms-like tyrosine kinase-1 may help stabilize the disease and prolong pregnancy. Interestingly, there are case reports suggesting that monoclonal antibody eculizumab (complement inhibitor) may have therapeutic potential. If new agents are discovered that are proven to be effective in preventing or treating preeclampsia, the potential to improve global maternal and perinatal health will be significant.
Collapse
|
4
|
Yu KH, Hung HY. Synthetic strategy and structure-activity relationship (SAR) studies of 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole (YC-1, Lificiguat): a review. RSC Adv 2021; 12:251-264. [PMID: 35424505 PMCID: PMC8978903 DOI: 10.1039/d1ra08120a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/14/2021] [Indexed: 01/04/2023] Open
Abstract
Since 1994, YC-1 (Lificiguat, 3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of hypoxia-inducible factor-1 (HIF-1) and NF-κB. Recently, Riociguat®, the first soluble guanylate cyclase (sGC) stimulator drug used to treat pulmonary hypertension and pulmonary arterial hypertension, was derived from the YC-1 structure. In this review, we aim to highlight the synthesis and structure–activity relationships in the development of YC-1 analogs and their possible indications. Since 1994, YC-1 (Lificiguat) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of HIF-1 and NF-κB.![]()
Collapse
Affiliation(s)
- Ko-Hua Yu
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| | - Hsin-Yi Hung
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| |
Collapse
|
5
|
Zhang J, Hua W, Zhao X, Yang F, Guo T, Zhang J, Zheng X, Liang W. Paeoniflorin alleviates endothelial dysfunction caused by overexpression of soluble fms-like tyrosine kinase 1 and soluble endoglin in preeclampsia via VEGFA upregulation. Biosci Biotechnol Biochem 2021; 85:814-823. [PMID: 33590855 DOI: 10.1093/bbb/zbaa106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/03/2020] [Indexed: 11/14/2022]
Abstract
This study assessed the protective effects of paeoniflorin against preeclampsia-related endothelial damage (ED). Human umbilical vein endothelial cells (HUVECs) isolated from healthy puerperae were identified by immunofluorescence assay. After paeoniflorin treatment, HUVECs were induced by soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sEng) to establish ED. Cell viability, migration, invasion, tube formation, and apoptosis were assessed by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) tetrazolium MTT assay, Scratch assay, Transwell assay, tube formation assay, and flow cytometry. VEGFA expression in HUVECs was analyzed by Western blot. HUVECs were successfully isolated and identified as Von Willebrand factor (vWF) positive. Individual treatment or cotreatment of sFlt-1 and sEng inhibited migration, invasion and tube formation, enhanced apoptosis, and decreased VEGFA expression in HUVECs. Paeoniflorin pretreatment partially reversed the effects delivered by cotreatment of sFlt-1 and sEng in HUVECs. Paeoniflorin alleviated preeclampsia-related ED caused by overexpression of sFlt-1 and sEng by upregulating VEGFA.
Collapse
Affiliation(s)
- Jin Zhang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wei Hua
- Reproductive Center, Xijing Hospital of Air Force Medical University, Xi'an City, Shan xi Province, China
| | - Xinyuan Zhao
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Fan Yang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Ting Guo
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jianhua Zhang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Xuerong Zheng
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wanqi Liang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
6
|
Duan J, Ma D, Wen X, Guo Q, Gao J, Zhang G, Xu K, Zhang L. Hydroxychloroquine prophylaxis for preeclampsia, hypertension and prematurity in pregnant patients with systemic lupus erythematosus: A meta-analysis. Lupus 2021; 30:1163-1174. [PMID: 33853420 DOI: 10.1177/09612033211007199] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES This meta-analysis aimed to evaluate the effectiveness of HCQ in improving the maternal and fetal outcomes in pregnancies with SLE. METHODS A literature search was conducted using PubMed, MEDLINE, EMBASE, and the Cochrane database for relevant English language articles, and Wanfang, CNKI and VIP for Chinese articles, from the databases' inception to April 30, 2020. These studies compared the maternal and/or fetal outcomes between pregnant patients with SLE who were administered HCQ during pregnancy (HCQ+ group) and those who were not administered HCQ (HCQ- group). Two investigators extracted the data and assessed the quality using the Newcastle-Ottawa Scale (NOS) and GRADE criteria independently. Odds ratio (OR) and 95% confidence intervals (CIs) were calculated. All statistical analyses were conducted using the Stata 12.0 software. RESULTS Nine studies involving 1132 pregnancies were included in the study (3 case controls, 2 prospective cohorts, 4 retrospective cohorts). Preeclampsia, gestational hypertension, and prematurity were significantly lower in the HCQ+ group than in the HCQ- group (OR 0.35, 95% CI 0.21-0.59), (OR 0.41, 95% CI 0.19-0.89) and (OR 0.55, 95% CI 0.36-0.86), respectively. There were no significant differences in the rates of HELLP Syndrome (OR 0.88, 95% CI 0.19-3.96), gestational diabetes (OR 2.3, 95% CI 0.44-12.12), thrombotic events (OR 0.26, 95% CI 0.05-1.51), spontaneous abortion (OR 1.77, 95% CI 0.96-3.26), premature rupture of membranes (OR 0.58, 95% CI 0.24-1.39), oligohydramnios (OR 0.90, 95% CI 0.38-2.14), live birth (OR 1.22, 95% CI 0.60-2.47), stillbirth (OR 1.00, 95% CI 0.50-2.00), congenital malformation (OR 0.53, 95% CI 0.14-2.04), low birth weight (OR 0.77, 95% CI 0.43-1.39), intrauterine distress (OR 1.07, 95% CI 0.41-2.76,), intrauterine growth restriction (OR 0.57, 95% CI 0.06-5.43), or five-minute APGAR score <7 (OR 0.72, 95% CI 0.20-2.58) between the two groups. CONCLUSIONS HCQ treatment during pregnancy could reduce the risk of preeclampsia, pregnancy hypertension and prematurity in SLE patients. The certainty of evidence is high but majority of the studies included are retrospective studies and not randomized controlled trials. Therefore, the multidisciplinary management of pregnant patients with SLE should promote HCQ use, irrespective of disease activity or severity.
Collapse
Affiliation(s)
- Jiaoniu Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China.,Tongji Shanxi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, China
| | - Xiaoting Wen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Qianyu Guo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China.,Tongji Shanxi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China.,Tongji Shanxi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, China
| | - Gailian Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
7
|
Godhamgaonkar AA, Sundrani DP, Joshi SR. Role of maternal nutrition and oxidative stress in placental telomere attrition in women with preeclampsia. Hypertens Pregnancy 2021; 40:63-74. [PMID: 33406938 DOI: 10.1080/10641955.2020.1869248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background:Maternal nutrition influences the growth and development of the fetus and influences pregnancy outcome. We have earlier demonstrated altered maternal nutrition and increased oxidative stress in women with preeclampsia. Oxidative stress is known to be associated with reduced telomere length and short telomere aggregates. Increased telomere attrition leads to increased cellular senescence and tissue ageing. Methods:The present review focuses on the role of maternal nutrition and oxidative stress in telomere attrition in preeclampsia. Results and Conclusion:Future studies need to examine the association between maternal nutritional status in early pregnancy, oxidative stress and telomere attrition in preeclampsia.
Collapse
Affiliation(s)
- Aditi A Godhamgaonkar
- Mother and Child Health, Interactive Research School of Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to Be University) , Pune, India
| | - Deepali P Sundrani
- Mother and Child Health, Interactive Research School of Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to Be University) , Pune, India
| | - Sadhana R Joshi
- Mother and Child Health, Interactive Research School of Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to Be University) , Pune, India
| |
Collapse
|
8
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Binder NK, Brownfoot FC, Beard S, Cannon P, Nguyen TV, Tong S, Kaitu'u-Lino TJ, Hannan NJ. Esomeprazole and sulfasalazine in combination additively reduce sFlt-1 secretion and diminish endothelial dysfunction: potential for a combination treatment for preeclampsia. Pregnancy Hypertens 2020; 22:86-92. [PMID: 32758704 DOI: 10.1016/j.preghy.2020.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Development and repurposing of therapies that show promise in the prevention or treatment of preeclampsia would be a major advance for the obstetrics field. We recently identified esomeprazole and sulfasalazine as potential candidates for the treatment of preeclampsia. Both reduce placental and endothelial secretion of sFlt-1 and sENG and mitigate endothelial dysfunction in vitro. Here we assessed whether esomeprazole and sulfasalazine in combination would additively attenuate the elevated release of anti-angiogenic factors and markers of endothelial dysfunction, key characteristics of preeclampsia. Primary placental tissue and cells, and primary endothelial cells were treated with esomeprazole and sulfasalazine alone and in combination. We assessed secretion of sFlt-1 and sENG and performed in vitro assays of endothelial dysfunction. Combining esomeprazole and sulfasalazine in lower concentrations caused an additive reduction in sFlt-1 secretion in primary cytotrophoblasts, placental explants and endothelial cells. No additive reduction was observed in sENG secretion when esomeprazole and sulfasalazine were combined. Together, esomeprazole and sulfasalazine additively reduced TNF-α-induced VCAM and ET-1 mRNA expression, and monocyte adhesion to endothelial cells. In conclusion, combining esomeprazole and sulfasalazine additively reduced secretion of sFlt-1 and markers of endothelial dysfunction. Combined administration of esomeprazole and sulfasalazine may provide a more effective treatment or prevention for preeclampsia compared to either as single agents.
Collapse
Affiliation(s)
- Natalie K Binder
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Therapeutics Discovery and Vascular Function, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Fiona C Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Sally Beard
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Therapeutics Discovery and Vascular Function, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Diagnostics Discovery and Reverse Translation, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Tuong V Nguyen
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Diagnostics Discovery and Reverse Translation, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Diagnostics Discovery and Reverse Translation, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Therapeutics Discovery and Vascular Function, Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Hospital for Women, Australia; Mercy Perinatal 163 Studley Rd, Heidelberg 3084, Victoria, Australia.
| |
Collapse
|
10
|
Rahman RA, Murthi P, Singh H, Gurungsinghe S, Leaw B, Mockler JC, Lim R, Wallace EM. Hydroxychloroquine Mitigates the Production of 8-Isoprostane and Improves Vascular Dysfunction: Implications for Treating Preeclampsia. Int J Mol Sci 2020; 21:E2504. [PMID: 32260307 PMCID: PMC7177667 DOI: 10.3390/ijms21072504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
In preeclampsia, widespread maternal endothelial dysfunction is often secondary to excessive generation of placental-derived anti-angiogenic factors, including soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), along with proinflammatory cytokines such as tumour necrosis factor-α (TNF-α) and activin A, understanding of which offers potential opportunities for the development of novel therapies. The antimalarial hydroxychloroquine is an anti-inflammatory drug improving endothelial homeostasis in lupus. It has not been explored as to whether it can improve placental and endothelial function in preeclampsia. In this in vitro study, term placental explants were used to assess the effects of hydroxychloroquine on placental production of sFlt-1, sEng, TNF-α, activin A, and 8-isoprostane after exposure to hypoxic injury or oxidative stress. Similarly, human umbilical vein endothelial cells (HUVECs) were used to assess the effects of hydroxychloroquine on in vitro markers of endothelial dysfunction. Hydroxychloroquine had no effect on the release of sFlt-1, sEng, TNF-α, activin A, or 8-isoprostane from placental explants exposed to hypoxic injury or oxidative stress. However, hydroxychloroquine mitigated TNF-α-induced HUVEC production of 8-isoprostane and Nicotinanamide adenine dinucleotide phosphate (NADPH) oxidase expression. Hydroxychloroquine also mitigated TNF-α and preeclamptic serum-induced HUVEC monolayer permeability and rescued the loss of zona occludens protein zona occludens 1 (ZO-1). Although hydroxychloroquine had no apparent effects on trophoblast function, it may be a useful endothelial protectant in women presenting with preeclampsia.
Collapse
Affiliation(s)
- Rahana Abd Rahman
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia; (S.G.); (J.C.M.); (R.L.)
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
- Department of Obstetrics and Gynaecology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur 56000, Malaysia
| | - Padma Murthi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Harmeet Singh
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
| | - Seshini Gurungsinghe
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia; (S.G.); (J.C.M.); (R.L.)
| | - Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
| | - Joanne C. Mockler
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia; (S.G.); (J.C.M.); (R.L.)
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia; (S.G.); (J.C.M.); (R.L.)
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
| | - Euan M. Wallace
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, Victoria 3168, Australia; (S.G.); (J.C.M.); (R.L.)
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; (H.S.); (B.L.)
| |
Collapse
|
11
|
Hitzerd E, Broekhuizen M, Neuman RI, Colafella KMM, Merkus D, Schoenmakers S, Simons SHP, Reiss IKM, Danser AHJ. Human Placental Vascular Reactivity in Health and Disease: Implications for the Treatment of Pre-eclampsia. Curr Pharm Des 2020; 25:505-527. [PMID: 30950346 DOI: 10.2174/1381612825666190405145228] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
Abstract
Adequate development of the placenta is essential for optimal pregnancy outcome. Pre-eclampsia (PE) is increasingly recognized to be a consequence of placental dysfunction and can cause serious maternal and fetal complications during pregnancy. Furthermore, PE increases the risk of neonatal problems and has been shown to be a risk factor for cardiovascular disease of the mother later in life. Currently, there is no adequate treatment for PE, mainly because its multifactorial pathophysiology remains incompletely understood. It originates in early pregnancy with abnormal placentation and involves a cascade of dysregulated systems in the placental vasculature. To investigate therapeutic strategies it is essential to understand the regulation of vascular reactivity and remodeling of blood vessels in the placenta. Techniques using human tissue such as the ex vivo placental perfusion model provide insight in the vasoactive profile of the placenta, and are essential to study the effects of drugs on the fetal vasculature. This approach highlights the different pathways that are involved in the vascular regulation of the human placenta, changes that occur during PE and the importance of focusing on restoring these dysfunctional systems when studying treatment strategies for PE.
Collapse
Affiliation(s)
- Emilie Hitzerd
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Michelle Broekhuizen
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Cardiology; Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Rugina I Neuman
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Gynecology and Obstetrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Katrina M Mirabito Colafella
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Melbourne, Australia
| | - Daphne Merkus
- Department of Cardiology; Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sam Schoenmakers
- Department of Gynecology and Obstetrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sinno H P Simons
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine; Division of Pharmacology and Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
12
|
Xiao S, Li Q, Hu L, Yu Z, Yang J, Chang Q, Chen Z, Hu G. Soluble Guanylate Cyclase Stimulators and Activators: Where are We and Where to Go? Mini Rev Med Chem 2019; 19:1544-1557. [PMID: 31362687 DOI: 10.2174/1389557519666190730110600] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/05/2019] [Accepted: 04/20/2019] [Indexed: 02/04/2023]
Abstract
Soluble Guanylate Cyclase (sGC) is the intracellular receptor of Nitric Oxide (NO). The activation of sGC results in the conversion of Guanosine Triphosphate (GTP) to the secondary messenger cyclic Guanosine Monophosphate (cGMP). cGMP modulates a series of downstream cascades through activating a variety of effectors, such as Phosphodiesterase (PDE), Protein Kinase G (PKG) and Cyclic Nucleotide-Gated Ion Channels (CNG). NO-sGC-cGMP pathway plays significant roles in various physiological processes, including platelet aggregation, smooth muscle relaxation and neurotransmitter delivery. With the approval of an sGC stimulator Riociguat for the treatment of Pulmonary Arterial Hypertension (PAH), the enthusiasm in the discovery of sGC modulators continues for broad clinical applications. Notably, through activating the NO-sGC-cGMP pathway, sGC stimulator and activator potentiate for the treatment of various diseases, such as PAH, Heart Failure (HF), Diabetic Nephropathy (DN), Systemic Sclerosis (SS), fibrosis as well as other diseases including Sickle Cell Disease (SCD) and Central Nervous System (CNS) disease. Here, we review the preclinical and clinical studies of sGC stimulator and activator in recent years and prospect for the development of sGC modulators in the near future.
Collapse
Affiliation(s)
- Sijia Xiao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Zutao Yu
- Department of Chemistry, Graduate School of Science Kyoto University Kitashirakawa- Oiwakecho, Sakyo-Ku, kyoto, Japan
| | - Jie Yang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Qi Chang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| |
Collapse
|
13
|
Kose S, Tuna G, Nuriyeva G, Altunyurt S, Islekel GH, Doğan OE. A prospective cohort study on the prediction of the diagnosis-to-delivery time in preeclamptic pregnancies: should the sFlt-1/PlGF ratio be added to routine evaluations? Arch Gynecol Obstet 2018; 298:911-920. [DOI: 10.1007/s00404-018-4903-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 09/06/2018] [Indexed: 11/24/2022]
|
14
|
Valero L, Alhareth K, Gil S, Lecarpentier E, Tsatsaris V, Mignet N, Fournier T, Andrieux K. Nanomedicine as a potential approach to empower the new strategies for the treatment of preeclampsia. Drug Discov Today 2018; 23:1099-1107. [DOI: 10.1016/j.drudis.2018.01.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/28/2017] [Accepted: 01/24/2018] [Indexed: 01/27/2023]
|
15
|
Kaitu’u-Lino TJ, Brownfoot FC, Beard S, Cannon P, Hastie R, Nguyen TV, Binder NK, Tong S, Hannan NJ. Combining metformin and esomeprazole is additive in reducing sFlt-1 secretion and decreasing endothelial dysfunction - implications for treating preeclampsia. PLoS One 2018; 13:e0188845. [PMID: 29466360 PMCID: PMC5821305 DOI: 10.1371/journal.pone.0188845] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/14/2017] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION The discovery of new treatments that prevent or treat preeclampsia would be a major advance. Antiangiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sENG) are secreted in excess from the placenta, causing hypertension, endothelial dysfunction, and multiorgan injury. We recently identified metformin and esomeprazole as potential treatments for preeclampsia. Both reduce placental and endothelial secretion of sFlt-1 and soluble endoglin, and reduce endothelial dysfunction. OBJECTIVES We set out to assess whether combining metformin and esomeprazole would additively reduce sFlt-1 and soluble endoglin secretion and reduce endothelial dysfunction (verses drug alone). Metformin and esomeprazole were added to primary placental cells and tissues, and endothelial cells and their effects on sFlt-1 and soluble endoglin secretion were assessed in vitro. Tumor necrosis factor-α (TNF-α) was added to endothelial cells to induce dysfunction in vitro. We examined the ability of metformin + esomeprazole to rescue TNF-α induced vascular cell adhesion molecule-1 (VCAM-1) and Endothelin-1 (ET-1) expression, leukocyte adhesion (markers of endothelial dysfunction). RESULTS Combining metformin and esomeprazole was additive at reducing sFlt-1 secretion and expression of sFlt-1 e15a mRNA isoform in primary cytotrophoblast, placental explants and endothelial cells. In contrast, no additive reduction in sENG was observed with combined metformin and esomeprazole. The low-dose combination of metformin + esomeprazole additively reduced TNF-α-induced VCAM-1 mRNA, but not VCAM-1 protein expression. There was no additive reduction when combining metformin and esomeprazole on TNF-α induced PBMC adhesion to endothelial cells. However, combining metformin and esomeprazole additively reduced ET-1 mRNA expression. CONCLUSIONS In conclusion combining metformin and esomeprazole additively reduced secretion of sFlt-1, and markers of endothelial dysfunction. The combination of metformin and esomeprazole may provide a more effective treatment or prevention for preeclampsia compared to either as single agents.
Collapse
Affiliation(s)
- Tu’uhevaha J. Kaitu’u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Fiona C. Brownfoot
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Sally Beard
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Roxanne Hastie
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong V. Nguyen
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natalie K. Binder
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natalie J. Hannan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne and Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- * E-mail:
| |
Collapse
|
16
|
Palmer KR, Tong S, Kaitu'u-Lino TJ. Placental-specific sFLT-1: role in pre-eclamptic pathophysiology and its translational possibilities for clinical prediction and diagnosis. Mol Hum Reprod 2018; 23:69-78. [PMID: 27986932 DOI: 10.1093/molehr/gaw077] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/09/2016] [Indexed: 11/12/2022] Open
Abstract
Pre-eclampsia is a common obstetric complication globally responsible for a significant burden of maternal and perinatal morbidity and mortality. Central to its pathophysiology is the anti-angiogenic protein, soluble fms-like tyrosine kinase-1 (sFLT-1). sFLT-1 is released from a range of tissues into the circulation, where it antagonizes the activity of vascular endothelial growth factor and placental growth factor leading to endothelial dysfunction. It is this widespread endothelial dysfunction that produces the clinical features of pre-eclampsia including hypertension and proteinuria. There are multiple splice variants of sFLT-1. One, known as sFLT-1 e15a, evolved quite recently and is only present in humans and higher order primates. This sFLT-1 variant is also the main sFLT-1 secreted from the placenta. Recent work has shown that sFLT-1 e15a is significantly elevated in the placenta and circulation of women with pre-eclampsia. It is also biologically active, capable of causing endothelial dysfunction and the end-organ dysfunction seen in pre-eclampsia. Indeed, the over-expression of sFLT-1 e15a in mice recapitulates the pre-eclamptic phenotype in pregnancy. Therefore, here we propose that sFLT-1 e15a may be the sFLT-1 variant primarily responsible for pre-eclampsia, a uniquely human disease. Furthermore, this placental-specific sFLT-1 variant provides promise for use as an accurate biomarker in the prediction or diagnosis of pre-eclampsia.
Collapse
Affiliation(s)
- K R Palmer
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Rd, Clayton, 3168 Victoria, Australia.,Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - S Tong
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - T J Kaitu'u-Lino
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| |
Collapse
|
17
|
Zhang Y, Zhao HJ, Xia XR, Diao FY, Ma X, Wang J, Gao L, Liu J, Gao C, Cui YG, Liu JY. Hypoxia-induced and HIF1α-VEGF-mediated tight junction dysfunction in choriocarcinoma cells: Implications for preeclampsia. Clin Chim Acta 2017; 489:203-211. [PMID: 29223764 DOI: 10.1016/j.cca.2017.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/22/2017] [Accepted: 12/05/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Accumulated data indicate that placental hypoxia is implicated in the pathogenesis of preeclampsia (PE). Tight junction (TJ) is important structure that sustains normal placental barrier function, its dysregulation under hypoxia has been observed. This study was designed to explore hypoxia-induced TJ dysfunction in trophoblast cells and its possible involvement in PE pathophysiology. METHODS Choriocarcinoma cells were grown in a monolayer and treated with cobalt chloride (CoCl2) to induce hypoxia. TJ architecture was assessed using transmission electron microscopy, and locations of TJ proteins were determined by immunofluorescence. TJ functions were assessed by transepithelial electrical resistance (TER) and increased cell paracellular permeability (CPP), and the expression of TJ-related proteins, HIF-1α and VEGF was measured. RESULTS The TJ functions of trophoblast cells were significantly altered by hypoxia; TER decreased and CPP increased in a time- and concentration-dependent manner. Significant alterations in TJ protein expression and increases in HIF1α and VEGF expression were observed in hypoxic cells, and these effects were attenuated by pretreatment with YC-1. Moreover, corresponding changes in TJ protein expression were also detected in preeclamptic placentas. CONCLUSION These data demonstrate that trophoblast cells undergo significant changes in TJ protein expression under hypoxic conditions and highlight the potential significance of the HIF1α-VEGF axis in the regulation of TJ structure and function in the preeclamptic placenta.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hai-Jun Zhao
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Reproductive Medicine, Central Hospital of Handan City, Handan, Hebei 956000, China
| | - Xin-Ru Xia
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Fei-Yang Diao
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiang Ma
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Wang
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jie Liu
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chao Gao
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu-Gui Cui
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jia-Yin Liu
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
18
|
Abstract
Placental dysfunction underlies major obstetric diseases such as pre-eclampsia and fetal growth restriction (FGR). Whilst there has been a little progress in prophylaxis, there are still no treatments for placental dysfunction in normal obstetric practice. However, a combination of increasingly well-described in vitro systems for studying the human placenta, together with the availability of more appropriate animal models of pre-eclampsia and FGR, has facilitated a recent surge in work aimed at repurposing drugs and therapies, developed for other conditions, as treatments for placental dysfunction. This review: (1) highlights potential candidate drug targets in the placenta - effectors of improved uteroplacental blood flow, anti-oxidants, heme oxygenase induction, inhibition of HIF, induction of cholesterol synthesis pathways, increasing insulin-like growth factor II availability; (2) proposes an experimental pathway for taking a potential drug or treatment for placental dysfunction from concept through to early phase clinical trials, utilizing techniques for studying the human placenta in vitro and small animal models, particularly the mouse, for in vivo studies; (3) describes the data underpinning sildenafil citrate and adenovirus expressing vascular endothelial growth as potential treatments for placental dysfunction and summarizes recent research on other potential treatments. The importance of sharing information from such studies even when no effect is found, or there is an adverse outcome, is highlighted. Finally, the use of adenoviral vectors or nanoparticle carriers coated with homing peptides to selectively target drugs to the placenta is highlighted: such delivery systems could improve efficacy and reduce the side effects of treating the dysfunctional placenta.
Collapse
Affiliation(s)
- Colin P Sibley
- Maternal and Fetal Health Research CentreDivision of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- St Mary's HospitalCentral Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
19
|
Li M, Jia Y, Ling Y, Chen Y, Zhang L, Luo D, Lai L, Guo M, Zhang D, Ren M, Xu H, Kuang H. Reduced expression of follicle stimulating hormone receptor mRNA and protein in pregnancies complicated by pre-eclampsia. Mol Med Rep 2017; 16:367-372. [DOI: 10.3892/mmr.2017.6599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 03/16/2017] [Indexed: 11/06/2022] Open
|
20
|
Hannan NJ, Brownfoot FC, Cannon P, Deo M, Beard S, Nguyen TV, Palmer KR, Tong S, Kaitu'u-Lino TJ. Resveratrol inhibits release of soluble fms-like tyrosine kinase (sFlt-1) and soluble endoglin and improves vascular dysfunction - implications as a preeclampsia treatment. Sci Rep 2017; 7:1819. [PMID: 28500309 PMCID: PMC5431923 DOI: 10.1038/s41598-017-01993-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 04/06/2017] [Indexed: 01/24/2023] Open
Abstract
Preeclampsia is a disease of pregnancy associated with placental oxidative stress, inflammation and elevated release of anti-angiogenic factors sFlt-1 and soluble endoglin. These placental factors cause generalized maternal endothelial dysfunction. There are no treatments to halt disease progression; delivery is the only cure. Resveratrol modulates pathways involved in inflammation and oxidative stress and may offer a potential therapeutic for preeclampsia. Resveratrol reduced sFlt-1, sFlt-1 e15a and soluble endoglin secretion from primary trophoblasts and HUVECs and reduced mRNA expression of pro-inflammatory molecules NFκB, IL-6 and IL-1β in trophoblasts. IL-6, IL-1β and TNFα secretion were also significantly reduced. In HUVECs, resveratrol significantly increased mRNA of anti-oxidant enzymes HO-1, NQO1, GCLC and TXN but did not significantly alter HO-1 protein expression, whilst reducing HO-1 protein in trophoblast. Endothelial dysfunction was induced in HUVECs using TNFα, increasing expression of cell adhesion molecule VCAM1 and adhesion of peripheral blood mononuclear cells, both of which were increased further by resveratrol. In contrast, resveratrol significantly reduced TNFα-induced Endothelin-1 (a vasoconstrictor) and significantly increased the phosphorylation of endothelial nitric oxide synthase (eNOS). In summary, resveratrol decreases secretion of anti-angiogenic factors however its effects on the endothelium are mixed. Overall, it may have potential as a treatment for preeclampsia.
Collapse
Affiliation(s)
- Natalie J Hannan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Fiona C Brownfoot
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Minh Deo
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Sally Beard
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Tuong V Nguyen
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Kirsten R Palmer
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Rd, Clayton, 3168, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Rd, Heidelberg, 3084, Victoria, Australia.
| |
Collapse
|
21
|
|
22
|
Onda K, Tong S, Beard S, Binder N, Muto M, Senadheera SN, Parry L, Dilworth M, Renshall L, Brownfoot F, Hastie R, Tuohey L, Palmer K, Hirano T, Ikawa M, Kaitu'u-Lino T, Hannan NJ. Proton Pump Inhibitors Decrease Soluble fms-Like Tyrosine Kinase-1 and Soluble Endoglin Secretion, Decrease Hypertension, and Rescue Endothelial Dysfunction. Hypertension 2017; 69:457-468. [PMID: 28115513 DOI: 10.1161/hypertensionaha.116.08408] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 09/28/2016] [Accepted: 12/12/2016] [Indexed: 01/30/2023]
Abstract
Preeclampsia is a severe complication of pregnancy. Antiangiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin are secreted in excess from the placenta, causing hypertension, endothelial dysfunction, and multiorgan injury. Oxidative stress and vascular inflammation exacerbate the endothelial injury. A drug that can block these pathophysiological steps would be an attractive treatment option. Proton pump inhibitors (PPIs) are safe in pregnancy where they are prescribed for gastric reflux. We performed functional studies on primary human tissues and animal models to examine the effects of PPIs on sFlt-1 and soluble endoglin secretion, vessel dilatation, blood pressure, and endothelial dysfunction. PPIs decreased sFlt-1 and soluble endoglin secretion from trophoblast, placental explants from preeclamptic pregnancies, and endothelial cells. They also mitigated tumor necrosis factor-α-induced endothelial dysfunction: PPIs blocked endothelial vascular cell adhesion molecule-1 expression, leukocyte adhesion to endothelium, and disruption of endothelial tube formation. PPIs decreased endothelin-1 secretion and enhanced endothelial cell migration. Interestingly, the PPI esomeprazole vasodilated maternal blood vessels from normal pregnancies and cases of preterm preeclampsia, but its vasodilatory effects were lost when the vessels were denuded of their endothelium. Esomeprazole decreased blood pressure in a transgenic mouse model where human sFlt-1 was overexpressed in placenta. PPIs upregulated endogenous antioxidant defenses and decreased cytokine secretion from placental tissue and endothelial cells. We have found that PPIs decrease sFlt-1 and soluble endoglin secretion and endothelial dysfunction, dilate blood vessels, decrease blood pressure, and have antioxidant and anti-inflammatory properties. They have therapeutic potential for preeclampsia and other diseases where endothelial dysfunction is involved.
Collapse
Affiliation(s)
- Kenji Onda
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Stephen Tong
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.).
| | - Sally Beard
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Natalie Binder
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Masanaga Muto
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Sevvandi N Senadheera
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Laura Parry
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Mark Dilworth
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Lewis Renshall
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Fiona Brownfoot
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Roxanne Hastie
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Laura Tuohey
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Kirsten Palmer
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Toshihiko Hirano
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Masahito Ikawa
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Tu'uhevaha Kaitu'u-Lino
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| | - Natalie J Hannan
- From the Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (K.O., S.T., S.B., N.B., F.B., R.H., L.T., K.P., T.K.-L., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan (K.O., T.H.); Research Institute for Microbial Diseases, Osaka University, Japan (M.M., M.I.); School of Biosciences, University of Melbourne, Parkville, Victoria, Australia (S.N.S., L.P.); Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, United Kingdom (M.D., L.R.); St Mary's Hospital, Central Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, United Kingdom (M.D., L.R.)
| |
Collapse
|
23
|
Hastie R, Tong S, Hannan NJ, Brownfoot F, Cannon P, Kaitu'u-Lino TJ. Epidermal Growth Factor Rescues Endothelial Dysfunction in Primary Human Tissues In Vitro. Reprod Sci 2016; 24:1245-1252. [PMID: 27920343 DOI: 10.1177/1933719116681516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy, responsible for over 60 000 maternal deaths annually. Endothelial dysfunction is a central aspect to its pathophysiology, and currently, no medical therapeutic is available for its treatment. In this study, we aim to investigate the effect of epidermal growth factor (EGF) on endothelial dysfunction using primary human tissues. We performed a number of in vitro assays that mimic the vascular endothelial dysfunction that occurs in preeclampsia. Epidermal growth factor reduced the expression of vascular cell adhesion molecule-1, a marker of endothelial dysfunction, after insult with tumor necrosis factor α (TNF-α) or serum from women with preeclampsia. Additionally, after TNF-α insult, EGF reduced tube disruption and the adhesion of monocytes to primary human umbilical vein endothelial cells (HUVECs). Our findings suggest that EGF reduces endothelial dysfunction in primary HUVECs. Epidermal growth factor may have potential as a novel peptide treatment for preeclampsia and other diseases where there is endothelial dysfunction.
Collapse
Affiliation(s)
- Roxanne Hastie
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen Tong
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| | - Natalie J Hannan
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| | - Fiona Brownfoot
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| | - Ping Cannon
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- 1 Department of Obstetrics and Gynaecology, Translational Obstetrics Group, Mercy Hospital for Women, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Mooney SS, Lee RM, Tong S, Brownfoot FC. Expectant management of severe preterm preeclampsia: a comparison of maternal and fetal indications for delivery. J Matern Fetal Neonatal Med 2016; 29:3821-6. [DOI: 10.3109/14767058.2016.1147555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Samantha S. Mooney
- Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Heidelberg, Victoria, Australia and
| | - Rilka M. Lee
- Department of Obstetrics and Gynaecology, Mercy Hospital for Women, Heidelberg, Victoria, Australia and
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Fiona C. Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
25
|
Metformin as a prevention and treatment for preeclampsia: effects on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion and endothelial dysfunction. Am J Obstet Gynecol 2016; 214:356.e1-356.e15. [PMID: 26721779 DOI: 10.1016/j.ajog.2015.12.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Preeclampsia is associated with placental ischemia/hypoxia and secretion of soluble fms-like tyrosine kinase 1 and soluble endoglin into the maternal circulation. This causes widespread endothelial dysfunction that manifests clinically as hypertension and multisystem organ injury. Recently, small molecule inhibitors of hypoxic inducible factor 1α have been found to reduce soluble fms-like tyrosine kinase 1 and soluble endoglin secretion. However, their safety profile in pregnancy is unknown. Metformin is safe in pregnancy and is also reported to inhibit hypoxic inducible factor 1α by reducing mitochondrial electron transport chain activity. OBJECTIVE The purposes of this study were to determine (1) the effects of metformin on placental soluble fms-like tyrosine kinase 1 and soluble endoglin secretion, (2) to investigate whether the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion are regulated through the mitochondrial electron transport chain, and (3) to examine its effects on endothelial dysfunction, maternal blood vessel vasodilation, and angiogenesis. STUDY DESIGN We performed functional (in vitro and ex vivo) experiments using primary human tissues to examine the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from placenta, endothelial cells, and placental villous explants. We used succinate, mitochondrial complex II substrate, to examine whether the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin secretion were mediated through the mitochondria. We also isolated mitochondria from preterm preeclamptic placentas and gestationally matched control subjects and measured mitochondrial electron transport chain activity using kinetic spectrophotometric assays. Endothelial cells or whole maternal vessels were incubated with metformin to determine whether it rescued endothelial dysfunction induced by either tumor necrosis factor-α (to endothelial cells) or placenta villous explant-conditioned media (to whole vessels). Finally, we examined the effects of metformin on angiogenesis on maternal omental vessel explants. RESULTS Metformin reduced soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from primary endothelial cells, villous cytotrophoblast cells, and preterm preeclamptic placental villous explants. The reduction in soluble fms-like tyrosine kinase 1 and soluble endoglin secretion was rescued by coadministration of succinate, which suggests that the effects of metformin on soluble fms-like tyrosine kinase 1 and soluble endoglin were likely to be regulated at the level of the mitochondria. In addition, the mitochondrial electron transport chain inhibitors rotenone and antimycin reduced soluble fms-like tyrosine kinase 1 secretion, which further suggests that soluble fms-like tyrosine kinase 1 secretion is regulated through the mitochondria. Mitochondrial electron transport chain activity in preterm preeclamptic placentas was increased compared with gestation-matched control subjects. Metformin improved features of endothelial dysfunction relevant to preeclampsia. It reduced endothelial cell messenger RNA expression of vascular cell adhesion molecule 1 that was induced by tumor necrosis factor-α (vascular cell adhesion molecule 1 is an inflammatory adhesion molecule up-regulated with endothelial dysfunction and is increased in preeclampsia). Placental conditioned media impaired bradykinin-induced vasodilation; this effect was reversed by metformin. Metformin also improved whole blood vessel angiogenesis impaired by fms-like tyrosine kinase 1. CONCLUSION Metformin reduced soluble fms-like tyrosine kinase 1 and soluble endoglin secretion from primary human tissues, possibly by inhibiting the mitochondrial electron transport chain. The activity of the mitochondrial electron transport chain was increased in preterm preeclamptic placenta. Metformin reduced endothelial dysfunction, enhanced vasodilation in omental arteries, and induced angiogenesis. Metformin has potential to prevent or treat preeclampsia.
Collapse
|