1
|
Rahimi S, Rezvani N, Khazayel S, Jalilian N, Shakiba E, Khadir F, Yari K, Rahimi Z. The study of HMOX1 DNA methylation and gene expression and the diagnostic potential of miR-153-3p in preeclampsia. Epigenomics 2024; 16:389-401. [PMID: 38410927 DOI: 10.2217/epi-2023-0377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Background: The objective was to elucidate the potential epigenetic regulatory mechanism in HMOX1 expression in preeclampsia. Materials & methods: HMOX1 promoter DNA methylation was evaluated in the placental tissue and blood of preeclamptic and normotensive pregnant women. HMOX1 and miR-153-3p gene expression were assessed in placental tissue and peripheral blood mononuclear cells (PBMCs). Related microarray datasets in the Gene Expression Omnibus database were also analyzed. Results: In placental tissue, despite HMOX1 expression downregulation, there was no significant change in HMOX1 methylation. In PBMCs, there was no significant alteration in HMOX1 expression, while hypomethylation was observed in blood. The miR-153-3p expression increased in the placental tissue and in the PBMCs of preeclampsia. Conclusion: DNA methylation does not affect HMOX1 expression, while miR-153-3p might be a biomarker for preeclampsia.
Collapse
Affiliation(s)
- Somayeh Rahimi
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
| | - Nayebali Rezvani
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
| | - Saeed Khazayel
- Deputy of Research & Technology, Kermanshah University of Medical Sciences, Kermanshah, 67146-73159, Iran
| | - Nazanin Jalilian
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
| | - Ebrahim Shakiba
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
| | - Fatemeh Khadir
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 67155-1616, Iran
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, 67148-69914, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 67155-1616, Iran
| |
Collapse
|
2
|
Nguyen A, Cannon P, Kandel M, Nguyen TV, Baird L, Wong G, Hannan NJ, Tong S, Bartho L, Kaitu'u-Lino TJ. Cell surface associated protein mucin 15 (MUC15) is elevated in preeclampsia. Placenta 2023; 140:39-46. [PMID: 37531748 DOI: 10.1016/j.placenta.2023.07.013] [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: 04/16/2023] [Revised: 06/14/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Mucins are a family of proteins that protect the epithelium. A particular type of mucin, MUC15 is highly expressed in the placenta. This study aimed to characterise MUC15 in preeclampsia and investigate its role in placental stem cell biology. METHODS MUC15 mRNA and protein were measured in placentas from patients with early onset (<34 weeks' gestation) preeclampsia. Circulating serum MUC15 was measured via ELISA. MUC15 was localised in the placenta using in situ hybridisation. MUC15 mRNA expression was measured across differentiation of human trophoblast stem cells (hTSCs) to syncytiotrophoblast and extravillous trophoblasts. MUC15 was measured after syncytialised hTSCs were cultured in hypoxic (1% O2) and proinflammatory (TNF α, IL-6) conditions. MUC15 secretion was assessed when syncytialised hTSCs were treated with brefeldin A (impairs protein trafficking) and batimastat (inhibits matrix metalloproteinases). RESULTS MUC15 protein was significantly increased in the placenta (P = 0.0003, n = 32 vs n = 20 controls) and serum (P = 0.016, n = 32 vs n = 22 controls) of patients with preeclampsia, whilst MUC15 mRNA remained unchanged (n = 61 vs n = 18 controls). MUC15 mRNA (P = 0.005) and protein secretion (P = 0.006) increased following differentiation to syncytiotrophoblast cells. In situ hybridisation confirmed MUC15 localised to the syncytiotrophoblast cell within the placenta. Neither hypoxic or inflammatory conditions changed MUC15 mRNA expression or secretion. Brefeldin A treated hTSCs did not alter MUC15 secretion, whilst batimastat reduced MUC15 secretion (P = 0.044). CONCLUSIONS MUC15 is increased in early onset preeclampsia and is cleaved by matrix metalloproteinases. Increased MUC15 may reflect a protective mechanism associated with placental dysfunction. Further research will aid in confirming this.
Collapse
Affiliation(s)
- Anna Nguyen
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Ping Cannon
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Manju Kandel
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tuong-Vi Nguyen
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Lydia Baird
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Georgia Wong
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Natalie J Hannan
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Stephen Tong
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Lucy Bartho
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia.
| |
Collapse
|
3
|
Furuta A, Shima T, Yoshida-Kawaguchi M, Yamada K, Yasuda I, Tsuda S, Yamaki-Ushijima A, Yoneda S, Higashisaka K, Cheng SB, Matsumoto K, Tsutsumi Y, Sharma S, Saito S, Nakashima A. Chloroquine is a safe autophagy inhibitor for sustaining the expression of antioxidant enzymes in trophoblasts. J Reprod Immunol 2023; 155:103766. [PMID: 36470134 DOI: 10.1016/j.jri.2022.103766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/19/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Inhibition of autophagy contributes to the pathophysiology of preeclampsia. Although chloroquine (CHQ) is an autophagy inhibitor, it can reduce the occurrence of preeclampsia in women with systemic lupus erythematosus. To clarify this important clinical question, this study aimed to address the safety of CHQ in trophoblast cells from the viewpoint of homeostasis, in which the anti-oxidative stress (OS) response and autophagy are involved. We used Western blotting to evaluate the protein levels in the trophoblast cells. The expression levels of heme oxygenase-1 (HO-1), an anti-OS enzyme, mediate resistance to OS induced by hydrogen peroxide (H2O2) in trophoblast cell lines. Among the autophagy modulators, bafilomycin A1 (BAF), an autophagy inhibitor, but not autophagy activators, suppressed HO-1 expression in BeWo cells; CHQ did not suppress HO-1 expression in BeWo cells. To clarify the role of autophagy in HO-1 induction, we observed no difference in HO-1 induction by H2O2 between autophagy-normal and autophagy-deficient cells. As for the mechanism of HO-1 induction by OS, BAF suppressed HO-1 induction by downregulating the expression of neighbor of BRCA1 gene 1 (NBR1) in the selective p62-NBR1-nuclear factor erythroid 2-related factor 2 (Nrf2) autophagy pathway. CHQ did not inhibit HO-1 expression by sustaining NBR1 expression in human villous tissues compared to BAF treatment. In conclusion, CHQ is a safer medicine than BAF for sustaining NBR1, which resist against OS in trophoblasts by connecting selective autophagy and the anti-OS response.
Collapse
Affiliation(s)
- Atsushi Furuta
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Tomoko Shima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Mihoko Yoshida-Kawaguchi
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Kiyotaka Yamada
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Ippei Yasuda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Sayaka Tsuda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Akemi Yamaki-Ushijima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Satoshi Yoneda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Kazuma Higashisaka
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shi-Bin Cheng
- Departments of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yasuo Tsutsumi
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; The Center for Advanced Medical Engineering and Informatics, Osaka University, 1-6, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Surendra Sharma
- Departments of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Shigeru Saito
- University of Toyama, 3190 Gofuku, Toyama 9308555, Japan
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan.
| |
Collapse
|
4
|
Sun T, Cruz GI, Mousavi N, Marić I, Brewer A, Wong RJ, Aghaeepour N, Sayed N, Wu JC, Stevenson DK, Leonard SA, Gymrek M, Winn VD. HMOX1 Genetic Polymorphisms Display Ancestral Diversity and May Be Linked to Hypertensive Disorders in Pregnancy. Reprod Sci 2022; 29:3465-3476. [PMID: 35697922 PMCID: PMC9734242 DOI: 10.1007/s43032-022-01001-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/02/2022] [Indexed: 12/14/2022]
Abstract
Racial disparity exists for hypertensive disorders in pregnancy (HDP), which leads to disparate morbidity and mortality worldwide. The enzyme heme oxygenase-1 (HO-1) is encoded by HMOX1, which has genetic polymorphisms in its regulatory region that impact its expression and activity and have been associated with various diseases. However, studies of these genetic variants in HDP have been limited. The objective of this study was to examine HMOX1 as a potential genetic contributor of ancestral disparity seen in HDP. First, the 1000 Genomes Project (1 KG) phase 3 was utilized to compare the frequencies of alleles, genotypes, and estimated haplotypes of guanidine thymidine repeats (GTn; containing rs3074372) and A/T SNP (rs2071746) among females from five ancestral populations (Africa, the Americas, Europe, East Asia, and South Asia, N = 1271). Then, using genomic DNA from women with a history of HDP, we explored the possibility of HMOX1 variants predisposing women to HDP (N = 178) compared with an equivalent ancestral group from 1 KG (N = 263). Both HMOX1 variants were distributed differently across ancestries, with African women having a distinct distribution and an overall higher prevalence of the variants previously associated with lower HO-1 expression. The two HMOX1 variants display linkage disequilibrium in all but the African group, and within EUR cohort, LL and AA individuals have a higher prevalence in HDP. HMOX1 variants demonstrate ancestral differences that may contribute to racial disparity in HDP. Understanding maternal genetic contribution to HDP will help improve prediction and facilitate personalized approaches to care for HDP.
Collapse
Affiliation(s)
- Tianyanxin Sun
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanna I Cruz
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Mousavi
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alina Brewer
- Preeclampsia Foundation, Juneau Biosciences, LLC, Salt Lake City, UT, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nazish Sayed
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephanie A Leonard
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa Gymrek
- Department of Medicine, Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Zhang X, Chen Y, Sun D, Zhu X, Ying X, Yao Y, Fei W, Zheng C. Emerging pharmacologic interventions for pre-eclampsia treatment. Expert Opin Ther Targets 2022; 26:739-759. [PMID: 36223503 DOI: 10.1080/14728222.2022.2134779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pre-eclampsia is a serious pregnancy complication and a major global concern for the mortality of both mother and fetus. Existing symptomatic treatments do not delay disease progression; thus, timely delivery of the baby is the most effective measure. However, the risk of various maternal and fetal injuries remains. AREAS COVERED In this review, we summarize the potential strategies for pharmacologic interventions in pre-eclamptic therapy. Specifically, we discuss the pathophysiological process of various effective candidate therapeutics that act on potential pathways and molecular targets to inhibit key stages of the disease. We refer to this pathogenesis-focused drug discovery model as a pathogenesis-target-drug (P-T-D) strategy. Finally, we discuss the introduction of nanotechnologies to improve the safety and efficacy of therapeutics via their specific placental targeting ability and placental retention effects. EXPERT OPINION Despite the active development of novel pharmacological treatments based on our current knowledge of pre-eclamptic pathogenesis, investigations are still in the early phase. Thus, further exploration of the pathological mechanisms, integrated with the P-T-D strategy and novel nanosystems, could encourage the development of more effective and safer strategies. Such advances could lead to a shift from expectant management to mechanistic-based therapy for pre-eclampsia.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Dongli Sun
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xia Ying
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
6
|
Iacopucci APM, da Silva Pereira P, Pereira DA, Calmasini FB, Pittalà V, Reis LO, Burnett AL, Costa FF, Silva FH. Intravascular hemolysis leads to exaggerated corpus cavernosum relaxation: Implication for priapism in sickle cell disease. FASEB J 2022; 36:e22535. [PMID: 36070139 DOI: 10.1096/fj.202200867r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
Patients with sickle cell disease (SCD) display priapism. Clinical studies have shown a strong positive correlation between priapism and high levels of intravascular hemolysis in men with SCD. However, there are no experimental studies that show that intravascular hemolysis promotes alterations in erectile function. Therefore, we aimed to evaluate the corpus cavernosum smooth muscle relaxant function in a murine model that displays intravascular hemolysis induced by phenylhydrazine (PHZ), as well as the role of intravascular hemolysis in increasing the stress oxidative in the penis. Corpus cavernosum strips were dissected free and placed in organ baths. Acetylcholine and electrical field stimulation (EFS)-induced corpus cavernosum relaxations in vitro were obtained. Increased corpus cavernosum relaxant responses to acetylcholine and EFS were observed in the PHZ group. Protein expression of heme oxygenase-1 increased in the corpus cavernosum of the PHZ group, but PDE5 protein expression was not modified. Preincubation with the heme oxygenase inhibitor 1 J completely reversed the increased relaxant responses to acetylcholine and EFS in PHZ mice. Protein expression of NADPH oxidase subunit gp91phox, 3-nitrotyrosine, and 4-hydroxynonenal increased in the corpus cavernosum of the PHZ group, suggesting a state of oxidative stress. Basal cGMP production was lower in the PHZ group. Our results show that intravascular hemolysis promotes increased corpus cavernosum smooth muscle relaxation associated with increased HO-1 expression, as well as increased oxidative stress associated with upregulation of gp91phox expression. Moreover, our study supports clinical studies that point to a strong positive correlation between priapism and high levels of intravascular hemolysis in men with SCD.
Collapse
Affiliation(s)
- Ana Paula Magrini Iacopucci
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, Brazil
| | | | - Dalila Andrade Pereira
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, Brazil
| | - Fabiano Beraldi Calmasini
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, Brazil
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Arthur L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Fábio Henrique Silva
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, Brazil
| |
Collapse
|
7
|
Zhang T, Shen HH, Qin XY, Li MQ. The metabolic characteristic of decidual immune cells and their unique properties in pregnancy loss. Immunol Rev 2022; 308:168-186. [PMID: 35582842 DOI: 10.1111/imr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.
Collapse
Affiliation(s)
- Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue-Yun Qin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,Shanghai Medical School, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Mégier C, Peoc’h K, Puy V, Cordier AG. Iron Metabolism in Normal and Pathological Pregnancies and Fetal Consequences. Metabolites 2022; 12:metabo12020129. [PMID: 35208204 PMCID: PMC8876952 DOI: 10.3390/metabo12020129] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Iron is required for energy production, DNA synthesis, and cell proliferation, mainly as a component of the prosthetic group in hemoproteins and as part of iron-sulfur clusters. Iron is also a critical component of hemoglobin and plays an important role in oxygen delivery. Imbalances in iron metabolism negatively affect these vital functions. As the crucial barrier between the fetus and the mother, the placenta plays a pivotal role in iron metabolism during pregnancy. Iron deficiency affects 1.2 billion individuals worldwide. Pregnant women are at high risk of developing or worsening iron deficiency. On the contrary, in frequent hemoglobin diseases, such as sickle-cell disease and thalassemia, iron overload is observed. Both iron deficiency and iron overload can affect neonatal development. This review aims to provide an update on our current knowledge on iron and heme metabolism in normal and pathological pregnancies. The main molecular actors in human placental iron metabolism are described, focusing on the impact of iron deficiency and hemoglobin diseases on the placenta, together with normal metabolism. Then, we discuss data concerning iron metabolism in frequent pathological pregnancies to complete the picture, focusing on the most frequent diseases.
Collapse
Affiliation(s)
- Charles Mégier
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France;
| | - Katell Peoc’h
- Assistance Publique-Hôpitaux de Paris, Laboratoire de Biochimie Clinique, HUPNVS, Hôpital Beaujon, Clichy and Université de Paris, UFR de Médecine Xavier Bichat, INSERM U1149, F-75018 Paris, France;
| | - Vincent Puy
- Unité de biologie de la Reproduction CECOS, Hôpital Antoine Béclère, Université Paris Saclay, 92140 Clamart, France;
- Laboratoire de Développement des Gonades, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA, F-92265 Fontenay-aux-Roses, France
| | - Anne-Gaël Cordier
- INSERM, 3PHM, UMR-S1139, F-75006 Paris, France
- PremUp Foundation, F-75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie Obstétrique, Hôpital Antoine Béclère, Université Paris-Saclay, 92140 Clamart, France
- Correspondence: ; Tel.: +33-145374441; Fax: +33-45374366
| |
Collapse
|
9
|
Cluver CA, Hiscock R, Decloedt EH, Hall DR, Schell S, Mol BW, Brownfoot F, Kaitu'u-Lino TJ, Walker SP, Tong S. Use of metformin to prolong gestation in preterm pre-eclampsia: randomised, double blind, placebo controlled trial. BMJ 2021; 374:n2103. [PMID: 34551918 PMCID: PMC8457042 DOI: 10.1136/bmj.n2103] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate whether extended release metformin could be used to prolong gestation in women being expectantly managed for preterm pre-eclampsia. DESIGN Randomised, double blind, placebo controlled trial. SETTING Referral hospital in Cape Town, South Africa. PARTICIPANTS 180 women with preterm pre-eclampsia between 26+0 to 31+6 weeks' gestation undergoing expectant management: 90 were randomised to extended release metformin and 90 to placebo. INTERVENTION 3 g of oral extended release metformin or placebo daily, in divided doses, until delivery. MAIN OUTCOME MEASURE The primary outcome was prolongation of gestation. RESULTS Of 180 participants, one woman delivered before taking any trial drug. The median time from randomisation to delivery was 17.7 days (interquartile range 5.4-29.4 days; n=89) in the metformin arm and 10.1 (3.7-24.1; n=90) days in the placebo arm, a median difference of 7.6 days (geometric mean ratio 1.39, 95% confidence interval 0.99 to 1.95; P=0.057). Among those who continued to take the trial drug at any dose, the median prolongation of gestation in the metformin arm was 17.5 (interquartile range 5.4-28.7; n=76) days compared with 7.9 (3.0-22.2; n=74) days in the placebo arm, a median difference of 9.6 days (geometric mean ratio 1.67, 95% confidence interval 1.16 to 2.42). Among those who took the full dosage, the median prolongation of gestation in the metformin arm was 16.3 (interquartile range 4.8-28.8; n=40) days compared with 4.8 (2.5-15.4; n=61) days in the placebo arm, a median difference of 11.5 days (geometric mean ratio 1.85, 95% confidence interval 1.14 to 2.88). Composite maternal, fetal, and neonatal outcomes and circulating concentrations of soluble fms-like tyrosine kinase-1, placental growth factor, and soluble endoglin did not differ. In the metformin arm, birth weight increased non-significantly and length of stay decreased in the neonatal nursery. No serious adverse events related to trial drugs were observed, although diarrhoea was more common in the metformin arm. CONCLUSIONS This trial suggests that extended release metformin can prolong gestation in women with preterm pre-eclampsia, although further trials are needed. It provides proof of concept that treatment of preterm pre-eclampsia is possible. TRIAL REGISTRATION Pan African Clinical Trial Registry PACTR201608001752102 https://pactr.samrc.ac.za/.
Collapse
Affiliation(s)
- Catherine A Cluver
- Department of Obstetrics and Gynaecology, Stellenbosch University, Tygerberg Hospital, 7505, Cape Town, South Africa
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Richard Hiscock
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Eric H Decloedt
- Department of Medicine, Division of Clinical Pharmacology, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - David R Hall
- Department of Obstetrics and Gynaecology, Stellenbosch University, Tygerberg Hospital, 7505, Cape Town, South Africa
| | - Sonja Schell
- Department of Obstetrics and Gynaecology, Stellenbosch University, Tygerberg Hospital, 7505, Cape Town, South Africa
| | - Ben W Mol
- Department of Obstetrics and Gynaecology, Monash School of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Fiona Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Susan P Walker
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
10
|
Meyer N, Langwisch S, Scharm M, Zenclussen AC. Using ultrasound to define the time point of intrauterine growth retardation in a mouse model of heme oxygenase-1 deficiency†. Biol Reprod 2021; 103:126-134. [PMID: 32342097 DOI: 10.1093/biolre/ioaa057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/09/2019] [Accepted: 04/27/2020] [Indexed: 12/17/2022] Open
Abstract
The enzyme heme oxygenase-1 (HO-1), encoded by the HMOX1 gene, mediates heme catabolism by cleaving free heme. We have previously revealed the importance of HO-1 in pregnancy. Here, we determined the impact of maternal or paternal HO-1 deficiency on fetal growth and placental parameters throughout gestation. We mated Hmox1-sufficient (WT), partial (HET)-, or total (KO)-deficient BALB/c female mice with Hmox1-WT or -KO BALB/c males and performed ultrasound analysis to monitor placental and fetal growth. Doppler measurements were used to determine maternal blood flow parameters. Offspring weights and feto-placental indices (FPI) were also determined. We found a significantly increased number of underdeveloped fetuses at gd10 in HET females that were mated with WT males compared with WT × WT pairings. At the same gestational age, underdeveloped placentas could be detected in HET females mated with KO males. Many fetuses from the KO × KO combination died in utero between gd12 and gd14. At gd14, abnormal placental parameters were found in surviving fetuses, which had significant reduced weights. Moreover, only 3.11% female and 5.33% male KO pups resulted from 10 HET × HET breeding pairs over 1 year. Our results show that HO-1 from both maternal and paternal origins is important for proper placental and fetal growth. Placental growth restriction and occurrence of abortions in mice that were partially or totally deficient in HO-1 were recorded in vivo from gd10 onwards. Future studies will focus on elucidating the cellular and molecular mechanisms behind these observations.
Collapse
Affiliation(s)
- Nicole Meyer
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Stefanie Langwisch
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Scharm
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
11
|
Chen S, Yin Q, Hu H, Chen Q, Huang Q, Zhong M. AOPPs induce HTR-8/SVneo cell apoptosis by downregulating the Nrf-2/ARE/HO-1 anti-oxidative pathway: Potential implications for preeclampsia. Placenta 2021; 112:1-8. [PMID: 34237527 DOI: 10.1016/j.placenta.2021.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Advanced oxidation protein products (AOPPs), which are novel markers of oxidant-mediated protein damage, are prevalent in numerous diseases. We previously demonstrated that AOPPs act as a new class of pathogenic mediators in preeclampsia by causing trophoblast damage and dysfunction. Herein, we explored whether AOPPs could regulate the Nrf-2/ARE/HO-1 anti-oxidative pathway to facilitate the progression of preeclampsia. METHODS To investigate the pathophysiology of preeclampsia, we evaluated the effects of AOPPs on trophoblast damage, apoptotic proteins, and Nrf-2/ARE/HO-1 anti-oxidative pathway expression, as well as their underlying mechanisms. RESULTS AOPPs directly increased the expression of apoptotic proteins and significantly inhibited the expression of Nrf-2/ARE/HO-1 pathway in trophoblasts. Nrf-2 silencing aggravated the AOPPs-induced cell apoptosis in vitro by activating p53 and caspase cascade, whereas Nrf-2 overexpression had the opposite effect. Moreover, Nrf-2 exerted cytoprotective effects by increasing HO-1. DISCUSSION These findings suggest that AOPPs induce trophoblast apoptosis by triggering p53 and caspase activation via inhibition of the Nrf-2/ARE/HO-1 anti-oxidative pathway. Hence, Nrf-2/ARE/HO-1 pathway activation plays a protective role in AOPPs-induced cell apoptosis; thus, holding potential as a therapeutic target against preeclampsia.
Collapse
Affiliation(s)
- Shuying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qitao Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Saraf SL. The vasculopathic cord between pre-eclampsia and kidney function in sickle cell disease. Br J Haematol 2021; 194:947-949. [PMID: 34121175 DOI: 10.1111/bjh.17574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 11/30/2022]
|
13
|
Ozen M, Zhao H, Kalish F, Yang Y, Jantzie LL, Wong RJ, Stevenson DK. Inflammation-induced alterations in maternal-fetal Heme Oxygenase (HO) are associated with sustained innate immune cell dysregulation in mouse offspring. PLoS One 2021; 16:e0252642. [PMID: 34086785 PMCID: PMC8177474 DOI: 10.1371/journal.pone.0252642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/19/2021] [Indexed: 12/28/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an evolutionarily conserved stress response enzyme and important in pregnancy maintenance, fetal and neonatal outcomes, and a variety of pathologic conditions. Here, we investigated the effects of an exposure to systemic inflammation late in gestation [embryonic day (E)15.5] on wild-type (Wt) and HO-1 heterozygous (Het, HO-1+/-) mothers, fetuses, and offspring. We show that alterations in fetal liver and spleen HO homeostasis during inflammation late in gestation can lead to a sustained dysregulation of innate immune cell populations and intracellular myeloid HO-1 expression in the spleen through young adolescence [postnatal day 25] in mice.
Collapse
Affiliation(s)
- Maide Ozen
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| | - Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Flora Kalish
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yang Yang
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lauren L. Jantzie
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ronald J. Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - David K. Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
14
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|
15
|
Zhao L, Wang X, Zhang X, Liu X, Ma N, Zhang Y, Zhang S. Therapeutic strategies for acute intermittent porphyria. Intractable Rare Dis Res 2020; 9:205-216. [PMID: 33139979 PMCID: PMC7586882 DOI: 10.5582/irdr.2020.03089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Acute intermittent porphyria (AIP) is an autosomal dominant disease caused by mutations in porphobilinogen deaminase (PBGD), the third enzyme of the heme synthesis pathway. Symptoms of AIP usually manifest as intermittent acute attacks with occasional neuropsychiatric crises. The management of AIP includes treatment of acute attacks, prevention of attacks, long-term monitoring and treatment of chronic complications. Intravenous injection of heme is the most effective method of treating acute attacks. Carbohydrate loading is used when heme is unavailable or in the event of mild attacks. Symptomatic treatment is also needed during attacks. Prevention of attacks includes eliminating precipitating factors, heme prophylaxis and liver transplantation. New treatment options include givosiran (siRNA) to down-regulate ALA synthase-1 (ALAS1) and the messenger RNA of PBGD (PBGD mRNA) delivered to the liver cells of patients with AIP. Long-term monitoring of chronic complications includes regular liver-kidney function and hepatocellular carcinoma (HCC) screening.
Collapse
Affiliation(s)
- Lanlan Zhao
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinyang Wang
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoning Zhang
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiantao Liu
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ningzhen Ma
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiran Zhang
- School of First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Songyun Zhang
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Address correspondence to:Songyun Zhang, Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China. E-mail:
| |
Collapse
|
16
|
Rezai H, Ahmad S, Alzahrani FA, Sanchez-Aranguren L, Dias IH, Agrawal S, Sparatore A, Wang K, Ahmed A. MZe786, a hydrogen sulfide-releasing aspirin prevents preeclampsia in heme oxygenase-1 haplodeficient pregnancy under high soluble flt-1 environment. Redox Biol 2020; 38:101768. [PMID: 33137710 PMCID: PMC7610044 DOI: 10.1016/j.redox.2020.101768] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/01/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023] Open
Abstract
Preeclampsia affects one in twelve of the 130 million pregnancies a year. The lack of an effective therapeutic to prevent or treat it is responsible for an annual global cost burden of 100 billion US dollars. Preeclampsia also affects these women later in life as it is a recognised risk factor for cardiovascular disease, stroke and vascular dementia. Our laboratory demonstrated that preeclampsia is associated with high soluble fms-like tyrosine kinase 1 (sFlt-1) and low heme oxygenase-1 (HO1/Hmox1) expression. Here we sought to determine the therapeutic value of a novel H2S-releasing aspirin (MZe786) in HO-1 haploid deficient (Hmox1+/−) pregnant mice in a high sFlt-1 environment. Pregnant Hmox1+/− mice were injected with adenovirus encoding sFlt-1 or control virus at gestation day E11.5. Subsequently, Hmox1+/− dams were treated daily with a number of treatment regimens until E17.5, when maternal and fetal outcomes were assessed. Here we show that HO-1 compromised mice in a high sFlt-1 environment during pregnancy exhibit severe preeclampsia signs and a reduction in antioxidant genes. MZe786 ameliorates preeclampsia by reducing hypertension and renal damage possibly by stimulating antioxidant genes. MZe786 also improved fetal outcome in comparison with aspirin alone and appears to be a better therapeutic agent at preventing preeclampsia than aspirin alone. Partial loss of heme oxygenase-1 under high soluble Flt-1 causes severe preeclampsia compared to high sFlt-1 alone. MZe786, hydrogen sulfide releasing aspirin prevents preeclampsia by suppressing maternal hypertension and kidney injury. MZe786 is able to rescue pregnancy and improves fetal outcome despite the persistent high levels of sFlt-1. MZe786 is a superior therapeutic candidate than aspirin in preventing preeclampsia.
Collapse
Affiliation(s)
- Homira Rezai
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom
| | - Shakil Ahmad
- Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom
| | - Faisal A Alzahrani
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Department of Biochemistry, ESC Research Unit, Faculty of Science, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Lissette Sanchez-Aranguren
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom
| | - Irundika Hk Dias
- Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom
| | - Swati Agrawal
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Department of Maternal Fetal Medicine, Mt Sinai Hospital, University of Toronto, Toronto, Canada
| | - Anna Sparatore
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Keqing Wang
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom
| | - Asif Ahmed
- Mirzyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Holt Street, Birmingham, B7 4BB, United Kingdom; Aston Medical Research Institute, Aston Medical School, Birmingham, United Kingdom; Department of Biochemistry, ESC Research Unit, Faculty of Science, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia; President's Office, University of Southampton, University Road, Southampton, SO17 1BJ, UK.
| |
Collapse
|
17
|
MZe786 Rescues Cardiac Mitochondrial Activity in High sFlt-1 and Low HO-1 Environment. Antioxidants (Basel) 2020; 9:antiox9070598. [PMID: 32660064 PMCID: PMC7402164 DOI: 10.3390/antiox9070598] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Hypertensive disorder in pregnancy is a major cause of maternal and perinatal mortality worldwide. Women who have had preeclampsia are at three to four times higher risk in later life of developing high blood pressure and heart disease. Soluble Flt-1 (sFlt-1) is elevated in preeclampsia and may remain high postpartum in women with a history of preeclampsia. Heme oxygenase-1 (Hmox1/HO-1) exerts protective effects against oxidative stimuli and is compromised in the placenta of pregnant women with preeclampsia. We hypothesized that sFlt-1 inhibits cardiac mitochondrial activity in HO-1 deficient mice. HO-1 haplo-insufficient mice (Hmox1+/−) were injected with adenovirus encoding sFlt-1 (Ad-sFlt-1) or control virus (Ad-CMV). Subsequently, they were treated daily with either placebo or MZe786 for six days, when the heart tissue was harvested to assess cardiac mitochondrial activity. Here, we show that the loss of HO-1 disturbed cardiac mitochondrial respiration and reduced mitochondrial biogenesis. The overexpression of sFlt-1 resulted in the inhibition of the cardiac mitochondrial activity in Hmox1+/− mice. The present study demonstrates that the hydrogen sulfide (H2S) releasing molecule, MZe786, rescues mitochondrial activity by stimulating cardiac mitochondrial biogenesis and antioxidant defense in Hmox1−/− mice and in Hmox1+/− mice exposed to a high sFlt-1 environment.
Collapse
|
18
|
HMOX1 is partly responsible for phenotypic and functional abnormalities in mesenchymal stem cells/stromal cells from placenta of preeclampsia (PE) patients. Stem Cell Res Ther 2020; 11:30. [PMID: 31964423 PMCID: PMC6975087 DOI: 10.1186/s13287-020-1557-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 01/12/2023] Open
Abstract
Background Preeclampsia is a common obstetric syndrome affecting women in their first pregnancy and characterized by hypertension and proteinuria, which appears after 20 weeks of gestation. It is characterized by high blood pressure and occasional damage to another organ system most often the liver and kidneys. Currently, the etiology and pathogenesis of this syndrome are not fully understood. Since mesenchymal stem cells/stromal cells (MSCs) are intimately associated with endothelial cells that line vessel walls in the decidua they may play some role in the pathogenesis of this syndrome. In this study, we have partly, unveiled the mechanism of preeclampsia pathogenesis at the stem cells level. Methods We have isolated and characterized MSCs from decidua basalis of preeclampsia placenta (PE-DBMSCs) and showed their decreased functionality in terms of proliferation, migration, adhesion and clone formation potential as compared to MSCs isolated from decidua region of normal placentae (DBMSCs). The cells were preconditioned with H2O2 and the functional characteristics were evaluated. Differentially expressed genes were analyzed using mass spectrometry. Immunoblotting confirmed the expression of these proteins. Results Pre-conditioning with H2O2 restored the functional outcome of PE-DBMSCs. Mass spectrometry (MS) analysis of differentially expressed proteins revealed HMOX1 as one of the major candidates missing in PE-DBMSCs. HMOX1 inhibition by tin protoporphyrin (SnPP) in normal DBMSCs resulted in a reduction in proliferation, migration, adhesion, and clone formation processes as compared to the untreated controls. mRNA and protein analyses of PE-DBMSCs preconditioned with H2O2 at lower doses showed upregulation of HMOX1 expression. Conclusions We hereby show for the first time that loss of function of stem cells/stromal cells isolated from the patients with preeclampsia may contribute towards the disease exacerbation. Our results suggest that HMOX1 may be partially responsible for the loss of functionality in PE-DBMSCs and contribute significantly towards the pathophysiology of preeclampsia. However, further investigation is required to decipher its exact role in the development and onset of the disorder.
Collapse
|
19
|
Miya M, Okamoto A, Nikaido T, Tachimoto-Kawaguchi R, Tanaka T. Immunohistochemical localization of heme oxygenase-1 and bilirubin/biopyrrin of heme metabolites as antioxidants in human placenta with preeclampsia. Hypertens Pregnancy 2019; 39:33-42. [DOI: 10.1080/10641955.2019.1688069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Michiko Miya
- Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Aikou Okamoto
- Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | - Tadao Tanaka
- Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Pulgar VM, Yasuda M, Gan L, Desnick RJ, Bonkovsky HL. Sex differences in vascular reactivity in mesenteric arteries from a mouse model of acute intermittent porphyria. Mol Genet Metab 2019; 128:376-381. [PMID: 30639047 PMCID: PMC6612470 DOI: 10.1016/j.ymgme.2019.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/08/2018] [Accepted: 01/05/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Acute intermittent porphyria (AIP) results from a partial deficiency of porphobilinogen deaminase (PBGD). Symptomatic AIP patients, most of whom are women, experience acute attacks characterized by severe abdominal pain and abrupt increases in blood pressure. Here, we characterized the reactivity of mesenteric arteries from male and female AIP mice with ~30% of normal PBGD activity and wild type C57BL/6 mice. METHODS An acute porphyric attack was induced in AIP mice by treatment with phenobarbital. Vascular responses to K+, phenylephrine (PE), acetylcholine (ACh), and hemin were determined (Wire Multi Myograph). RESULTS Maximal contraction to PE was increased in arteries from male and female AIP mice (p < .05) during an induced attack of acute porphyria. Female AIP arteries had increased sensitivity to PE (p < .05) even after nitric oxide (NO) blockade with Nω-nitro-L-arginine methyl ester (L-NAME) (p < .05). Maximal relaxation to ACh was similar in males and females with lower sensitivity in female AIP arteries (p < .05). Hemin induced greater relaxation in AIP arteries in both males and females (p < .05). SUMMARY/CONCLUSIONS Sex differences in this AIP mouse model include a pro-contractile response in females. These alterations may contribute to the increased blood pressure during an acute attack and provide a novel mechanism of action whereby heme ameliorates the attacks.
Collapse
Affiliation(s)
- Victor M Pulgar
- Department of Pharmaceutical Sciences, Campbell University, Buies Creek, NC, USA; Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Makiko Yasuda
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA.
| | - Lin Gan
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA.
| | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA.
| | - Herbert L Bonkovsky
- Section on Gastroenterology & Hepatology, Wake Forest University/NC Baptist Medical Center, Winston-Salem, NC, USA.
| |
Collapse
|
21
|
Fodor P, White B, Khan R. Inflammation-The role of ATP in pre-eclampsia. Microcirculation 2019; 27:e12585. [PMID: 31424615 DOI: 10.1111/micc.12585] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/23/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022]
Abstract
Sterile inflammation may be initiated by molecules in the host organism that signal "damage" or "danger" also known as danger-associated molecular pattern (DAMPs). In pre-eclampsia (PE), a variety of DAMPs may be involved in the etiology or exacerbation of the disorder. Adenosine 5'-triphosphate (ATP) is a key intracellular energy molecule as well as a ligand for purinergic receptors. In humans, under physiological conditions, extracellular ATP (eATP) levels are distinctly low, but can rise to several hundred fold when cells become injured, stressed, or even necrotic. This often initiates a sterile inflammatory response with eATP acting as a DAMP. Extracellular ATP and its derivative nucleotides synthetized by endonucleotidases exhibit many of their effects through purinergic receptors, via inflammatory cascades and the production of proinflammatory molecules. This is clearly seen in the P2X7 gated receptor, which is linked to release of cytokines of the interleukin-1 family. Considering its fundamental role in innate immunity, an imbalance of P2X7 receptor activation may lead to deleterious effects in the coordination of placental vessel tone via the synthesis of various proinflammatory cytokines. This review explores the implication of DAMPs, specifically ATP and uric acid in the inflammation associated with PE.
Collapse
Affiliation(s)
- Paul Fodor
- Division of Medical Science and Graduate Entry Medicine, School of Medicine, University of Nottingham, Medical School, Royal Derby Hospital Centre, Derby, UK
| | - Benjamin White
- Division of Medical Science and Graduate Entry Medicine, School of Medicine, University of Nottingham, Medical School, Royal Derby Hospital Centre, Derby, UK
| | - Raheela Khan
- Division of Medical Science and Graduate Entry Medicine, School of Medicine, University of Nottingham, Medical School, Royal Derby Hospital Centre, Derby, UK
| |
Collapse
|
22
|
Prieto J, Gonzalez-Aseguinolaza G. Acute Intermittent Porphyria: Novel Etiologic and Pathogenic Therapies Based on RNA Transfer to the Liver. Hepatology 2019; 70:1061-1063. [PMID: 31026336 DOI: 10.1002/hep.30678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Jesus Prieto
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research, CIMA, University of Navarra, IdisNA, Pamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research, CIMA, University of Navarra, IdisNA, Pamplona, Spain
| |
Collapse
|
23
|
Phipps EA, Thadhani R, Benzing T, Karumanchi SA. Pre-eclampsia: pathogenesis, novel diagnostics and therapies. Nat Rev Nephrol 2019; 15:275-289. [PMID: 30792480 PMCID: PMC6472952 DOI: 10.1038/s41581-019-0119-6] [Citation(s) in RCA: 576] [Impact Index Per Article: 115.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pre-eclampsia is a complication of pregnancy that is associated with substantial maternal and fetal morbidity and mortality. The disease presents with new-onset hypertension and often proteinuria in the mother, which can progress to multi-organ dysfunction, including hepatic, renal and cerebral disease, if the fetus and placenta are not delivered. Maternal endothelial dysfunction due to circulating factors of fetal origin from the placenta is a hallmark of pre-eclampsia. Risk factors for the disease include maternal comorbidities, such as chronic kidney disease, hypertension and obesity; a family history of pre-eclampsia, nulliparity or multiple pregnancies; and previous pre-eclampsia or intrauterine fetal growth restriction. In the past decade, the discovery and characterization of novel antiangiogenic pathways have been particularly impactful both in increasing understanding of the disease pathophysiology and in directing predictive and therapeutic efforts. In this Review, we discuss the pathogenic role of antiangiogenic proteins released by the placenta in the development of pre-eclampsia and review novel therapeutic strategies directed at restoring the angiogenic imbalance observed during pre-eclampsia. We also highlight other notable advances in the field, including the identification of long-term maternal and fetal risks conferred by pre-eclampsia.
Collapse
Affiliation(s)
- Elizabeth A Phipps
- Nephrology Division, Brigham and Women's Hospital, Boston, MA, USA
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Thadhani
- Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - S Ananth Karumanchi
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Nephrology Division, Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
24
|
Li J, Zhou J, Tian B, Chu Y, Zhang N, Hu X, Wan X, Ye Y. Activation of HO-1 protects placental cells function in oxidative stress via regulating ZO-1/occludin. Biochem Biophys Res Commun 2019; 511:903-909. [PMID: 30851935 DOI: 10.1016/j.bbrc.2019.02.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 02/27/2019] [Indexed: 12/20/2022]
Abstract
We previously confirmed that Nuclear factor erythroid 2-related factor-2 (Nrf2) and heme oxygenase (HO-1) play synergistic roles in the pathogenesis of preeclampsia. To further explore the function of HO-1 in the pathogenesis of preeclampsia, we established oxidative stress models respectively with human first-trimester trophoblast/simian virus (HTR8/SVneo) and human umbilical vein endothelial cells (HUVECs) and then assessed the effect of HO-1 on the two cell lines in oxidative stress conditions. The cell oxidative stress models were incubated with Hemin (an inducer of HO-1), then, the HTR8/SVneo cells were transfected by ZO-1 small interfering RNA (siRNA). The HTR-8/SVneo invasive abilities were detected, and the tube formation abilities of HUVECs were measured. HO-1 and tight junction proteins zonula occludens-1 (ZO-1) and occludin in the cells were detected. In both the trophoblastic and HUVEC oxidative stress models, HO-1、ZO-1 and occludin were increased incubated with Hemin. Meanwhile, HTR-8/SVneo cells incubated with Hemin showed increased invasion function against the destruction of hydrogen peroxide (H2O2). Similarly, the tube formation ability of HUVECs incubated with Hemin was increased. The above-mentioned effects were disappeared after HTR-8/SVneo cells were transfected by ZO-1 siRNA. These results suggest that HO-1 protects the function of placental cells in oxidative stress via regulating ZO-1/occludin.
Collapse
Affiliation(s)
- Jing Li
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Zhou
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baogang Tian
- Department of Radiology, Zhangqiu People's Hospital, Jinan, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyu Hu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Wan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanhua Ye
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
25
|
Malik A, Jee B, Gupta SK. Preeclampsia: Disease biology and burden, its management strategies with reference to India. Pregnancy Hypertens 2018; 15:23-31. [PMID: 30825923 DOI: 10.1016/j.preghy.2018.10.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 11/19/2022]
Abstract
Preeclampsia is the cause of significant maternal and fetal mortality and morbidity. It is characterized by new-onset hypertension and proteinuria after 20 weeks of gestation. Preeclamptic women and children born from preeclamptic pregnancies are at greater risk to develop severe cardiovascular complications and metabolic syndromes later in life. The incidence of preeclampsia is estimated to be seven times higher in developing countries as compared to the developed countries. This review summarizes the pathophysiology of preeclampsia, emerging new hypothesis of its origin, risk factors that make women susceptible to developing preeclampsia and the potential of various biomarkers being studied to predict preeclampsia. The health care of developing countries is continuously challenged by substantial burden of maternal and fetal mortality. India despite being a fast developing country, is still far behind in achieving the required maternal mortality rates as per Millennium Development Goals set by the World Health Organization. Further, this review discusses the prevalence of preeclampsia in India, health facilities to manage preeclampsia, current guidelines and protocols followed and government policies to combat this complication in Indian condition.
Collapse
Affiliation(s)
- Ankita Malik
- Reproductive Cell Biology Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067, India.
| | - Babban Jee
- Department of Health Research, Ministry of Health and Family Welfare, Government of India, New Delhi 110 001, India
| | - Satish Kumar Gupta
- Reproductive Cell Biology Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110 067, India.
| |
Collapse
|
26
|
Aldika Akbar MI, Herdiyantini M, Aryananda RA, CIninta N, Wardhana MP, Gumilar KE, Wicaksono B, Ernawati, Sulistyono A, Aditiawarman, Joewono HT, Nadir Abdullah, Dekker G, Dachlan EG, Dikman Angsar M. Serum heme oxygenase 1 (HO-1), soluble FMS like tyrosine kinase (sFlt-1) level, and neonatal outcome in early onset, late onset preeclampsia, and normal pregnancy. Hypertens Pregnancy 2018; 37:175-181. [DOI: 10.1080/10641955.2018.1494187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Muhammad Ilham Aldika Akbar
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Mita Herdiyantini
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Rozi Aditya Aryananda
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Nareswari CIninta
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Manggala Pasca Wardhana
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Khanisyah Erza Gumilar
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Budi Wicaksono
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Ernawati
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Agus Sulistyono
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Aditiawarman
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Hermanto Tri Joewono
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Nadir Abdullah
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Gustaaf Dekker
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
- Department Obstetrics & Gynecology, Lyell Mc Ewin Hospital, The University of Adelaide, Adelaide, Australia
| | - Erry Gumilar Dachlan
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Dikman Angsar
- Department Obstetrics & Gynecology, DR. Soetomo – UNAIR Hospital, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
27
|
Brook A, Hoaksey A, Gurung R, Yoong EEC, Sneyd R, Baynes GC, Bischof H, Jones S, Higgins LE, Jones C, Greenwood SL, Jones RL, Gram M, Lang I, Desoye G, Myers J, Schneider H, Hansson SR, Crocker IP, Brownbill P. Cell free hemoglobin in the fetoplacental circulation: a novel cause of fetal growth restriction? FASEB J 2018; 32:5436-5446. [PMID: 29723064 DOI: 10.1096/fj.201800264r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Cell free hemoglobin impairs vascular function and blood flow in adult cardiovascular disease. In this study, we investigated the hypothesis that free fetal hemoglobin (fHbF) compromises vascular integrity and function in the fetoplacental circulation, contributing to the increased vascular resistance associated with fetal growth restriction (FGR). Women with normal and FGR pregnancies were recruited and their placentas collected freshly postpartum. FGR fetal capillaries showed evidence of erythrocyte vascular packing and extravasation. Fetal cord blood fHbF levels were higher in FGR than in normal pregnancies ( P < 0.05) and the elevation of fHbF in relation to heme oxygenase-1 suggests a failure of expected catabolic compensation, which occurs in adults. During ex vivo placental perfusion, pathophysiological fHbF concentrations significantly increased fetal-side microcirculatory resistance ( P < 0.05). fHbF sequestered NO in acute and chronic exposure models ( P < 0.001), and fHbF-primed placental endothelial cells developed a proinflammatory phenotype, demonstrated by activation of NF-κB pathway, generation of IL-1α and TNF-α (both P < 0.05), uncontrolled angiogenesis, and disruption of endothelial cell flow alignment. Elevated fHbF contributes to increased fetoplacental vascular resistance and impaired endothelial protection. This unrecognized mechanism for fetal compromise offers a novel insight into FGR as well as a potential explanation for associated poor fetal outcomes such as fetal demise and stillbirth.-Brook, A., Hoaksey, A., Gurung, R., Yoong, E. E. C., Sneyd, R., Baynes, G. C., Bischof, H., Jones, S., Higgins, L. E., Jones, C., Greenwood, S. L., Jones, R. L., Gram, M., Lang, I., Desoye, G., Myers, J., Schneider, H., Hansson, S. R., Crocker, I. P., Brownbill, P. Cell free hemoglobin in the fetoplacental circulation: a novel cause of fetal growth restriction?
Collapse
Affiliation(s)
- Adam Brook
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Annie Hoaksey
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rekha Gurung
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Edward E C Yoong
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rosanna Sneyd
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Georgia C Baynes
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Helen Bischof
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sarah Jones
- School of Healthcare Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Lucy E Higgins
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Carolyn Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Magnus Gram
- Division of Infection Medicine, Lund University, Lund, Sweden
| | - Ingrid Lang
- Institute of Histology and Embryology, University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Clinical Obstetrics and Gynecology, University of Graz, Graz, Austria
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Henning Schneider
- Department of Obstetrics and Gynecology, Inselspital, University of Bern, Bern, Switzerland
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Lund University, Lund, Sweden
| | - Ian P Crocker
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Paul Brownbill
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
28
|
Circulating Heme Oxygenase-1: Not a Predictor of Preeclampsia but Highly Expressed in Pregnant Women Who Subsequently Develop Severe Preeclampsia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6035868. [PMID: 30363976 PMCID: PMC6186313 DOI: 10.1155/2018/6035868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/24/2018] [Accepted: 08/30/2018] [Indexed: 11/18/2022]
Abstract
Preeclampsia is the major cause of maternal and fetal deaths worldwide. Circulating biomarker concentrations to predict preeclampsia must be determined. Therefore, the objective was to evaluate heme oxygenase-1 (HO-1) concentration in both plasma and urine samples from pregnant women before the development of preeclampsia and to identify a potential biomarker for preeclampsia development. We performed a case-control study nested in a prospective study cohort at University Hospital of the Ribeirao Preto Medical School, University of São Paulo (HCFMRP-USP), Ribeirao Preto, Brazil. Of 1400 pregnant women evaluated at 20–25 weeks of gestation, 460 delivered in hospitals outside our institution. Of 940 pregnant women who completed the protocol, 30 developed preeclampsia (cases, 14 cases of severe preeclampsia and 16 cases of mild preeclampsia). Healthy pregnant women (controls, n = 90) were randomly selected from the remaining 910 participants. HO-1 concentration was evaluated in plasma/urine samples by using a commercial enzyme-linked immunosorbent assay kit. We found similar HO-1 levels in the plasma and urine for case and control groups. In the subgrouped preeclampsia, lower plasma HO-1 levels were found in mild compared with severe preeclampsia. We conclude that plasma HO-1 levels were not altered at 20–25 weeks of gestation before the manifestation of preeclampsia symptoms. Pregnant women who subsequently develop severe preeclampsia show higher expression of HO-1. This may be indicative of important underlying pathophysiologic mechanisms that differentiate between mild and severe preeclampsia and may possibly be related to a higher prooxidative status even before the development of clinical symptoms.
Collapse
|
29
|
Bakrania BA, Spradley FT, Satchell SC, Stec DE, Rimoldi JM, Gadepalli RSV, Granger JP. Heme oxygenase-1 is a potent inhibitor of placental ischemia-mediated endothelin-1 production in cultured human glomerular endothelial cells. Am J Physiol Regul Integr Comp Physiol 2018; 314:R427-R432. [PMID: 29212810 PMCID: PMC5899255 DOI: 10.1152/ajpregu.00370.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/07/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder of maternal hypertension and reduced renal hemodynamics linked to reduced endothelial function. Placental ischemia is thought to be the culprit of this disease, as it causes the release of factors like tumor necrosis factor (TNF)-α that induce vascular endothelin-1 (ET-1) production. Interestingly, placental ischemia-induced hypertension in rats [reduced uterine perfusion pressure (RUPP) model] is abolished by ETA receptor blockade, suggesting a critical role for ET-1. Although it has been found that systemic induction of heme oxygenase (HO)-1 is associated with reduced ET-1 production and attenuated hypertension, it is unclear whether HO-1 directly modulates the increased ET-1 response to placental factors. We tested the hypothesis that HO-1 or its metabolites inhibit ET-1 production in human glomerular endothelial cells induced by serum of RUPP rats or TNF-α. Serum (5%) from RUPP hypertensive (mean arterial blood pressure 119 ± 9 mmHg) vs. normotensive pregnant (NP, 101 ± 6 mmHg, P < 0.001) rats increased ET-1 production (RUPP 168.8 ± 18.1 pg/ml, NP 80.3 ± 22.7 pg/ml, P < 0.001, n = 12/group). HO-1 induction [25 µM cobalt photoporphyrin (CoPP)] abolished RUPP serum-induced ET-1 production (1.6 ± 0.8 pg/ml, P < 0.001), whereas bilirubin (10 µM) significantly attenuated ET-1 release (125.3 ± 5.2 pg/ml, P = 0.005). Furthermore, TNF-α-induced ET-1 production (TNF-α 31.0 ± 8.4 vs. untreated 7.5 ± 0.4 pg/ml, P < 0.001) was reduced by CoPP (1.5 ± 0.8 pg/ml, P < 0.001) and bilirubin (10.5 ± 4.3 pg/ml, P < 0.001). These results suggest that circulating factors released during placental ischemia target the maternal glomerular endothelium to increase ET-1, and that pharmacological induction of HO-1 or bilirubin could be a treatment strategy to block this prohypertensive pathway in preeclampsia.
Collapse
Affiliation(s)
- Bhavisha A Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Frank T Spradley
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Surgery, University of Mississippi Medical Center , Jackson, Mississippi
| | - Simon C Satchell
- School of Clinical Sciences, University of Bristol , Bristol , United Kingdom
| | - David E Stec
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - John M Rimoldi
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Rama S V Gadepalli
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
30
|
|
31
|
George EM, Cockrell K, Arany M, Stec DE, Rimoldi JM, Gadepalli RSV, Granger JP. Carbon Monoxide Releasing Molecules Blunt Placental Ischemia-Induced Hypertension. Am J Hypertens 2017; 30:931-937. [PMID: 28472389 DOI: 10.1093/ajh/hpx070] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/31/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Preeclampsia is a pregnancy complication which manifests as new-onset hypertension, proteinuria, and a spectrum of other symptoms. While the underlying causes are still a subject of much debate, it is commonly believed that placental ischemia is a central cause. The ischemic placenta secretes factors which are believed to be responsible for the maternal syndrome; most notably the anti-angiogenic protein soluble fms-like tyrosine kinase 1 (sFlt-1). We have reported that induction of the carbon monoxide (CO) producing protein heme oxygenase-1 restored angiogenic imbalance and reduced blood pressure in a rat model of placental ischemia, and that CO blocks hypoxia-induced sFlt-1 production from placental tissue in vitro. We therefore hypothesized that direct administration of CO by a CO-releasing molecule (CORM) would blunt the placental ischemia-induced increase in sFlt-1 and thus the hypertension characteristic of this model. METHODS We administered a soluble CO donor molecule (CORM-3) daily i.v. in control animals or those undergoing placental ischemia from GD14. Blood pressure and renal function were measured on GD19, and angiogenic markers measured by ELISA. RESULTS Interestingly, though we found that CORM administration significantly blunted the hypertensive response to placental ischemia, there was no concomitant normalization of sFlt-1 in either the placenta or maternal circulation. We did find, however, that CORM administration caused a significant increase in glomerular filtration rate, presumably by vasodilation of the renal arteries and increased renal plasma flow. CONCLUSIONS All in all these data suggest that administration of CO by CORMs do lower blood pressure during placental ischemia mechanisms independent of changes in angiogenic balance.
Collapse
Affiliation(s)
- Eric M George
- Departments of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kathy Cockrell
- Departments of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Marietta Arany
- Departments of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David E Stec
- Departments of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - John M Rimoldi
- Department of BioMolecular Sciences, The University of Mississippi, University, Mississippi, USA
| | - Rama SV Gadepalli
- Department of BioMolecular Sciences, The University of Mississippi, University, Mississippi, USA
| | - Joey P Granger
- Departments of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
32
|
Abstract
Systemic lupus erythematosus (lupus, SLE) is a chronic autoimmune disease characterized by the production of autoantibodies, which bind to antigens and are deposited within tissues to fix complement, resulting in widespread systemic inflammation. The studies presented herein are consistent with hyperpolarized, adenosine triphosphate (ATP)-deficient mitochondria being central to the disease process. These hyperpolarized mitochondria resist the depolarization required for activation-induced apoptosis. The mitochondrial ATP deficits add to this resistance to apoptosis and also reduce the macrophage energy that is needed to clear apoptotic bodies. In both cases, necrosis, the alternative pathway of cell death, results. Intracellular constituents spill into the blood and tissues, eliciting inflammatory responses directed at their removal. What results is “autoimmunity.” Ultraviolet (UV)-A1 photons have the capacity to remediate this aberrancy. Exogenous exposure to low-dose, full-body, UV-A1 radiation generates singlet oxygen. Singlet oxygen has two major palliative actions in patients with lupus and the UV-A1 photons themselves have several more. Singlet oxygen depolarizes the hyperpolarized mitochondrion, triggering non-ATP-dependent apoptosis that deters necrosis. Next, singlet oxygen activates the gene encoding heme oxygenase (HO-1), a major governor of systemic homeostasis. HO-1 catalyzes the degradation of the oxidant heme into biliverdin (converted to bilirubin), Fe, and carbon monoxide (CO), the first three of these exerting powerful antioxidant effects, and in conjunction with a fourth, CO, protecting against injury to the coronary arteries, the central nervous system, and the lungs. The UV-A1 photons themselves directly attenuate disease in lupus by reducing B cell activity, preventing the suppression of cell-mediated immunity, slowing an epigenetic progression toward SLE, and ameliorating discoid and subacute cutaneous lupus. Finally, a combination of these mechanisms reduces levels of anticardiolipin antibodies and protects during lupus pregnancy. Capping all of this is that UV-A1 irradiation is an essentially innocuous, highly manageable, and comfortable therapeutic agency.
Collapse
Affiliation(s)
- H McGrath
- Veterans Administration, New Orleans, LA, USA
| |
Collapse
|
33
|
Kadam L, Gomez-Lopez N, Mial TN, Kohan-Ghadr HR, Drewlo S. Rosiglitazone Regulates TLR4 and Rescues HO-1 and NRF2 Expression in Myometrial and Decidual Macrophages in Inflammation-Induced Preterm Birth. Reprod Sci 2017; 24:1590-1599. [PMID: 28322133 DOI: 10.1177/1933719117697128] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Elevated inflammation accounts for approximately 30% of preterm birth (PTB) cases. We previously reported that targeting the peroxisome proliferator-activated receptor gamma (PPARγ) pathway reduced the incidence of PTB in the mouse model of endotoxin-induced PTB. The PPARγ has proven anti-inflammatory functions and its activation via rosiglitazone significantly downregulated the systemic inflammatory response and reduced PTB and stillbirth rate by 30% and 41%, respectively, in our model. Oxidative stress is inseparable from inflammation, and rosiglitazone has a reported antioxidative activity. In the current study, we therefore aimed to evaluate whether rosiglitazone treatment had effects outside of inflammatory pathway, specifically on the antioxidation pathway in our model. METHODS Pregnant C57BL/6J mice (E16.5) were treated with phosphate-buffered saline (PBS), rosiglitazone (Rosi), lipopolysaccharide (LPS; 10µg in 200µL 1XPBS), or LPS + Rosi (6 hours after the LPS injection). The myometrial and decidual tissues were collected and processed for macrophage isolation using magnetic cell sorting and F4/80+ antibody. Expression levels of antioxidative factors- Nrf2 and Ho-1-along with the LPS receptor Tlr4 were quantified by quantitative polymerase chain reaction. The protein levels were assessed by immunofluorescence staining. RESULTS Both the decidual and myometrial macrophages from the LPS-treated animals showed significantly lowered expression of Ho-1 and Nrf2 and higher expression of Tlr4 when compared to the PBS control group. The macrophages from the animals in the LPS + Rosi group had significantly elevated expression of Ho-1 and Nrf2 and downregulated expression of Tlr4 when compared to the LPS group. CONCLUSION Rosiglitazone administration prevents PTB by downregulating inflammation and upregulating antioxidative response.
Collapse
Affiliation(s)
- Leena Kadam
- 1 Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA.,2 Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- 2 Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA.,3 Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Services, Detroit, MI, USA.,4 Department of Immunology and Microbiology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Tara N Mial
- 2 Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA.,3 Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Services, Detroit, MI, USA
| | - Hamid-Reza Kohan-Ghadr
- 2 Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Sascha Drewlo
- 2 Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| |
Collapse
|
34
|
Abstract
Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.
Collapse
Affiliation(s)
- Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Abolfazl Zarjou
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Anupam Agarwal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
35
|
Zhao S, Zhang Y, Gu Y, Lewis DF, Wang Y. Heme Oxygenase-1 Mediates Up-Regulation of Adhesion Molecule Expression Induced by Peroxynitrite in Endothelial Cells. ACTA ACUST UNITED AC 2016; 11:465-71. [PMID: 15458743 DOI: 10.1016/j.jsgi.2004.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Endothelial cell (EC) activation with up-regulation of cellular adhesion molecule (CAM) expression is a pathophysiologic feature in preeclampsia (PE). Enhanced peroxynitrite formation in the vasculature of women with PE was also reported. This study was to test whether EC oxidative stress induced by peroxynitrite could up-regulate EC CAM expression, and whether heme oxygenase-1 (HO-1) has protective effects on this peroxynitrite-induced cellular response. METHODS Confluent ECs were stimulated with 3-morpholinosydnonimine-HCl (SIN-1, a peroxynitrite generator) alone or combined with Mn(III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride (MnTMPyP, a peroxynitrite scavenger) up to 4 hours. EC surface protein expressions for ICAM, VCAM, P-selectin, and E-selectin were measured by colorimetric assay. ECs were also treated with Sn(IV) mesophorphyrin IX dichloride (SnMP, a HO-1 inhibitor) to determine if HO-1 was involved in the increased CAM expression in stressed cells. Protein and mRNA expressions for HO-1 were determined by Western blot analysis and reverse-transcriptase polymerase chain reaction (RT-PCR). Data are presented as the mean +/- SE and analyzed by analysis of variance (ANOVA). RESULTS Endothelial CAM expressions for VCAM, P-selectin, and E-selectin, but not ICAM, were significantly increased in SIN-1-treated ECs. Protein and mRNA expressions for HO-1 were also up-regulated in cells treated with SIN-1. MnTMPyP blocked both mRNA and protein expressions for HO-1, whereas SnMP only blocked HO-1 protein expression. Both MnTMPyP and SnMP abolished SIN-1-induced up-regulation of VCAM, P-selectin, and E-selectin expression in ECs. CONCLUSIONS Peroxynitrite-induced EC oxidative stress produces differential effects on CAM expression, which may be mediated by HO-1 regulation. Our results suggest that increased peroxynitrite formation in the maternal vasculature may contribute to the increased CAM expression and enhanced neutrophil-endothelial interaction associated with PE.
Collapse
Affiliation(s)
- Shuang Zhao
- Departments of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | | | |
Collapse
|
36
|
Phipps E, Prasanna D, Brima W, Jim B. Preeclampsia: Updates in Pathogenesis, Definitions, and Guidelines. Clin J Am Soc Nephrol 2016; 11:1102-1113. [PMID: 27094609 PMCID: PMC4891761 DOI: 10.2215/cjn.12081115] [Citation(s) in RCA: 350] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preeclampsia is becoming an increasingly common diagnosis in the developed world and remains a high cause of maternal and fetal morbidity and mortality in the developing world. Delay in childbearing in the developed world feeds into the risk factors associated with preeclampsia, which include older maternal age, obesity, and/or vascular diseases. Inadequate prenatal care partially explains the persistent high prevalence in the developing world. In this review, we begin by presenting the most recent concepts in the pathogenesis of preeclampsia. Upstream triggers of the well described angiogenic pathways, such as the heme oxygenase and hydrogen sulfide pathways, as well as the roles of autoantibodies, misfolded proteins, nitric oxide, and oxidative stress will be described. We also detail updated definitions, classification schema, and treatment targets of hypertensive disorders of pregnancy put forth by obstetric and hypertensive societies throughout the world. The shift has been made to view preeclampsia as a systemic disease with widespread endothelial damage and the potential to affect future cardiovascular diseases rather than a self-limited occurrence. At the very least, we now know that preeclampsia does not end with delivery of the placenta. We conclude by summarizing the latest strategies for prevention and treatment of preeclampsia. A better understanding of this entity will help in the care of at-risk women before delivery and for decades after.
Collapse
Affiliation(s)
- Elizabeth Phipps
- Department of Nephrology/Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York; and
| | - Devika Prasanna
- Department of Nephrology/Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York; and
| | - Wunnie Brima
- Department of Medicine, James J. Peters Veterans Affairs Medical Center, New York, New York
| | - Belinda Jim
- Department of Nephrology/Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York; and
| |
Collapse
|
37
|
Zheng Y, Zhao Y, Luo Q, Liu X, Liu X, Hu Y, Zou L. Edaravone protects against cobalt chloride-induced dysfunctions in apoptosis and invasion in trophoblast cells. Mol Reprod Dev 2016; 83:576-87. [PMID: 27128210 DOI: 10.1002/mrd.22652] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/24/2016] [Indexed: 12/24/2022]
Affiliation(s)
- YanFang Zheng
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - Yin Zhao
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - QingQing Luo
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - XiaoXia Liu
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - XiaoPing Liu
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - Ying Hu
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| | - Li Zou
- Department of Obstetrics and Gynecology; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei Province China
| |
Collapse
|
38
|
Ahmed A, Ramma W. Unravelling the theories of pre-eclampsia: are the protective pathways the new paradigm? Br J Pharmacol 2016; 172:1574-86. [PMID: 25303561 PMCID: PMC4354257 DOI: 10.1111/bph.12977] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 09/30/2014] [Accepted: 10/05/2014] [Indexed: 01/03/2023] Open
Abstract
Pre-eclampsia is a vascular disorder of pregnancy where anti-angiogenic factors, systemic inflammation and oxidative stress predominate, but none can claim to cause pre-eclampsia. This review provides an alternative to the ‘two-stage model’ of pre-eclampsia in which abnormal spiral arteries modification leads to placental hypoxia, oxidative stress and aberrant maternal systemic inflammation. Very high maternal soluble fms-like tyrosine kinase-1 (sFlt-1 also known as sVEGFR) and very low placenta growth factor (PlGF) are unique to pre-eclampsia; however, abnormal spiral arteries and excessive inflammation are also prevalent in other placental disorders. Metaphorically speaking, pregnancy can be viewed as a car with an accelerator and brakes, where inflammation, oxidative stress and an imbalance in the angiogenic milieu act as the ‘accelerator’. The ‘braking system’ includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-γ-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. The failure in these pathways (brakes) results in the pregnancy going out of control and the system crashing. Put simply, pre-eclampsia is an accelerator–brake defect disorder. CO and H2S hold great promise because of their unique ability to suppress the anti-angiogenic factors sFlt-1 and soluble endoglin as well as to promote PlGF and endothelial NOS activity. The key to finding a cure lies in the identification of cheap, safe and effective drugs that induce the braking system to keep the pregnancy vehicle on track past the finishing line.
Collapse
Affiliation(s)
- Asif Ahmed
- Vascular Therapeutics Unit, Aston Medical School, Aston University, Birmingham, UK
| | | |
Collapse
|
39
|
Ahmed A, Rezai H, Broadway-Stringer S. Evidence-Based Revised View of the Pathophysiology of Preeclampsia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 956:355-374. [PMID: 27873232 DOI: 10.1007/5584_2016_168] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Preeclampsia is a life-threatening vascular disorder of pregnancy due to a failing stressed placenta. Millions of women risk death to give birth each year and globally each year, almost 300,000 lose their life in this process and over 500,000 babies die as a consequence of preeclampsia. Despite decades of research, we lack pharmacological agents to treat it. Maternal endothelial oxidative stress is a central phenomenon responsible for the preeclampsia phenotype of high maternal blood pressure and proteinuria. In 1997, it was proposed that preeclampsia arises due to the loss of VEGF activity, possibly due to elevation in anti-angiogenic factor, soluble Flt-1 (sFlt-1). Researchers showed that high sFlt-1 and soluble endoglin (sEng) elicit the severe preeclampsia phenotype in pregnant rodents. We demonstrated that heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway prevents placental stress and suppresses sFlt-1 and sEng release. Likewise, hydrogen sulphide (H2S)/cystathionine-γ-lyase (Cth) systems limit sFlt-1 and sEng and protect against the preeclampsia phenotype in mice. Importantly, H2S restores placental vasculature, and in doing so improves lagging fetal growth. These molecules act as the inhibitor systems in pregnancy and when they fail, preeclampsia is triggered. In this review, we discuss what are the hypotheses and models for the pathophysiology of preeclampsia on the basis of Bradford Hill causation criteria for disease causation and how further in vivo experimentation is needed to establish 'proof of principle'. Hypotheses that fail to meet the Bradford Hill causation criteria include abnormal spiral artery remodelling and inflammation and should be considered associated or consequential to the disorder. In contrast, the protection against cellular stress hypothesis that states that the protective pathways mitigate cellular stress by limiting elevation of anti-angiogenic factors or oxidative stress and the subsequent clinical signs of preeclampsia appear to fulfil most of Bradford Hill causation criteria. Identifying the candidates on the roadmap to this pathway is essential in developing diagnostics and therapeutics to target the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Asif Ahmed
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK.
| | - Homira Rezai
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Sophie Broadway-Stringer
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| |
Collapse
|
40
|
Zhang D, Liu H, Zeng J, Miao X, Huang W, Chen H, Huang Y, Li Y, Ye D. Glucocorticoid exposure in early placentation induces preeclampsia in rats via interfering trophoblast development. Gen Comp Endocrinol 2016; 225:61-70. [PMID: 26407501 DOI: 10.1016/j.ygcen.2015.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/18/2015] [Accepted: 09/17/2015] [Indexed: 12/29/2022]
Abstract
In pregnancy, placenta can be exposed to glucocorticoids (GCs) via several ways, which may disturb placentation and adversely affect pregnancy. Preeclampsia (PE) is thought to be attributed, in part, to impaired trophoblast development. The purpose of the present study was to confirm that GC exposure in early placentation could lead to PE in rats, with the mechanisms involving dysregulated trophoblast development. In the study, pregnant rats were administered with 2.5mg/kg Dex subcutaneously once per day from gestational day 7 to 13. Maternal systolic blood pressure and urinary albumin were increased, while both fetus and placenta were restricted after GC exposure relative to the control group. GC exposure also contributed to placental abnormalities and renal impairment. Moreover, placental oxidative damage was increased along with placental hypoxia-inducible factor 1-alpha (HIF1A) overexpression after GC treatment. Mechanically, GC induced PE in rat partially through inhibiting trophoblast proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), which involved phospho-extracellular signal regulated kinase (p-ERK) downregulation. Furthermore, GC receptor was required for the inhibition of GC on trophoblast proliferation, migration, invasion and EMT in vitro. These findings suggest that GC exposure in early placentation could contribute to PE in pregnant rats, with the mechanisms involving inhibition of trophoblast proliferation, migration, invasion and EMT by GC.
Collapse
Affiliation(s)
- Dongxin Zhang
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China; Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Haojing Liu
- Department of Internal Medicine, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Ji Zeng
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Xili Miao
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Wei Huang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Yinping Huang
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325003, People's Republic of China
| | - Yongsheng Li
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Duyun Ye
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
41
|
Ornaghi S, Mueller M, Barnea ER, Paidas MJ. Thrombosis during pregnancy: Risks, prevention, and treatment for mother and fetus-harvesting the power of omic technology, biomarkers and in vitro or in vivo models to facilitate the treatment of thrombosis. ACTA ACUST UNITED AC 2015; 105:209-25. [DOI: 10.1002/bdrc.21103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sara Ornaghi
- Department of Obstetrics and Gynecology; University of Milan-Bicocca; Monza Italy
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
| | - Martin Mueller
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
- Department of Obstetrics and Gynecology; University Hospital Bern; Bern Switzerland
| | - Eytan R. Barnea
- Society for the Investigation of Early Pregnancy; Cherry Hill New Jersey
- BioIncept LLC; Cherry Hill New Jersey
| | - Michael J. Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
| |
Collapse
|
42
|
Tong S, Kaitu'u-Lino TJ, Onda K, Beard S, Hastie R, Binder NK, Cluver C, Tuohey L, Whitehead C, Brownfoot F, De Silva M, Hannan NJ. Heme Oxygenase-1 Is Not Decreased in Preeclamptic Placenta and Does Not Negatively Regulate Placental Soluble fms-Like Tyrosine Kinase-1 or Soluble Endoglin Secretion. Hypertension 2015; 66:1073-81. [PMID: 26324507 DOI: 10.1161/hypertensionaha.115.05847] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022]
Abstract
Elevated placental release of the antiangiogenic factors, soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sENG), is central to the pathophysiology of preeclampsia. It is widely accepted that heme oxygenase-1 (HO-1) is decreased in preeclamptic placenta and negatively regulates sFlt-1 and sENG production. We set out to verify these contentions. There was no difference in HO-1 mRNA or protein levels in preterm preeclamptic placentas (n=17) compared with gestationally matched controls (n=27). In silico analysis of microarray studies did not identify decreased placental HO-1 expression in preeclamptic placenta. Silencing HO-1 in primary trophoblasts did not affect sFlt-1 protein secretion after 24 or 48 hours. Silencing nuclear factor (erythroid-derived 2)-like 2 (transcription factor that upregulates HO-1) in trophoblasts also did not affect sFlt-1 secretion. Administering tin protoporphyrin IX dichloride (HO-1 inhibitor) or cobalt protoporphyrin (HO-1 inducer) into placental explants did not affect sFlt-1 or sENG secretion. Silencing HO-1 in 2 types of primary endothelial cells (human umbilical vein endothelial and uterine microvascular endothelial cells) significantly increased sFlt-1 secretion but not sENG secretion. However, HO-1 silencing selectively increased mRNA expression of sFlt-1 i13 (generically expressed sFlt-1 variant) but not of sFlt-1 e15a (sFlt-1 variant mainly expressed in placenta). Furthermore, adding tin protoporphyrin IX dichloride decreased sFlt-1, whereas adding HO-1 inducers (cobalt protoporphyrin, dimethyl fumarate, and rosiglitazone) either had no effect or increased sFlt-1 or sENG secretion (these trends are opposite to what is expected). We conclude that HO-1 expression is not decreased in preeclamptic placenta and HO-1 does not negatively regulate placental sFlt-1 and sENG secretion in placental or endothelial cells.
Collapse
Affiliation(s)
- Stephen Tong
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.).
| | - Tu'uhevaha J Kaitu'u-Lino
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Kenji Onda
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Sally Beard
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Roxanne Hastie
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Natalie K Binder
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Cathy Cluver
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Laura Tuohey
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Clare Whitehead
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Fiona Brownfoot
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Manarangi De Silva
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| | - Natalie J Hannan
- From the Translational Obstetrics Group, the Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia (S.T., T.J.K.-L., K.O., S.B., R.H., N.K.B., C.C., L.T., C.W., F.B., M.D.S., N.J.H.); Department of Clinical Pharmacology, Tokyo University of Pharmacy and Life Sciences, School of Pharmacy, Tokyo, Japan (K.O.); and Department of Obstetrics and Gynaecology, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South Africa (C.C.)
| |
Collapse
|
43
|
George EM, Stout JM, Stec DE, Granger JP. Heme oxygenase induction attenuates TNF-α-induced hypertension in pregnant rodents. Front Pharmacol 2015; 6:165. [PMID: 26347650 PMCID: PMC4538306 DOI: 10.3389/fphar.2015.00165] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/24/2015] [Indexed: 11/13/2022] Open
Abstract
Pre-eclampsia is a hypertensive disorder of pregnancy initiated by placental insufficiency and chronic ischemia. In response, several pathways activated in the placenta are responsible for the maternal syndrome, including increased production of the anti-angiogenic protein, sFlt-1, and inflammatory cytokines, especially tumor necrosis factor-alpha (TNF-α). Previous studies have demonstrated that heme oxygenase (HO) induction can block TNF-α pathways in vitro and attenuate placental ischemia-induced sFlt-1 in vivo. Here, we investigated whether HO-1 induction could attenuate TNF-α-induced hypertension in pregnant rats. In response to TNF-α infusion (100 ng/day i.p.), maternal mean arterial pressure (MAP) increased vs. control animals (104 ± 3 vs. 119 ± 3 mmHg). HO-1 induction had no effect in control animals, but significantly decreased MAP in TNF-α-infused animals (108 ± 2 mmHg). Placental vascular endothelial growth factor (VEGF) was decreased in response to TNF-α infusion (92 ± 4 vs. 76 ± 2 pg/mg). Placental sFlt-1 was increased by TNF-α infusion (758 ± 45 vs. 936 ± 46 pg/mg, p < 0.05), which trended to normalization by HO-1 induction (779 ± 98 pg/mg). In contrast, HO-1 induction had no significant effect on placental VEGF in TNF-α-infused animals. Taken together, these data suggest that one of the key mechanisms by which HO exerts cytoprotective actions in the placenta during inflammation due to chronic ischemia is through suppression of sFlt-1. Further work elucidating the bioactive metabolites of HO-1 in innate inflammatory responses to placental ischemia is warranted.
Collapse
Affiliation(s)
- Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, MS, USA ; Department of Biochemistry, University of Mississippi Medical Center , Jackson, MS, USA
| | - Jacob M Stout
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, MS, USA
| | - David E Stec
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, MS, USA
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, MS, USA
| |
Collapse
|
44
|
Ha JG, Li L, Lee DH, Na SH, Ha KS, Kim YM, Ko JH, Hwang JY. The Effects of Heme Oxygenase By-Products on the Proliferation and Invasion of HUVECs, HTR-8/SVneo Cells, 3A(tPA 30-1) Cells, and HESCs Under Varying Oxygen Concentrations. Reprod Sci 2015; 22:1530-8. [DOI: 10.1177/1933719115589415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Joong Gyu Ha
- Department of Obstetrics and Gynecology, Eulji University Hospital, Daejeon, Korea
| | - Lan Li
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Dong heon Lee
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Sung hun Na
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Kwon Soo Ha
- Department of Molecular and Cell Biochemistry, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Young-Myeong Kim
- Department of Molecular and Cell Biochemistry, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jung Hwa Ko
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jong Yun Hwang
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
45
|
Solano ME, Kowal MK, O'Rourke GE, Horst AK, Modest K, Plösch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC. Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 2015; 125:1726-38. [PMID: 25774501 DOI: 10.1172/jci68140] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies in Western societies. IUGR is a strong predictor of reduced short-term neonatal survival and impairs long-term health in children. Placental insufficiency is often associated with IUGR; however, the molecular mechanisms involved in the pathogenesis of placental insufficiency and IUGR are largely unknown. Here, we developed a mouse model of fetal-growth restriction and placental insufficiency that is induced by a midgestational stress challenge. Compared with control animals, pregnant dams subjected to gestational stress exhibited reduced progesterone levels and placental heme oxygenase 1 (Hmox1) expression and increased methylation at distinct regions of the placental Hmox1 promoter. These stress-triggered changes were accompanied by an altered CD8+ T cell response, as evidenced by a reduction of tolerogenic CD8+CD122+ T cells and an increase of cytotoxic CD8+ T cells. Using progesterone receptor- or Hmox1-deficient mice, we identified progesterone as an upstream modulator of placental Hmox1 expression. Supplementation of progesterone or depletion of CD8+ T cells revealed that progesterone suppresses CD8+ T cell cytotoxicity, whereas the generation of CD8+CD122+ T cells is supported by Hmox1 and ameliorates fetal-growth restriction in Hmox1 deficiency. These observations in mice could promote the identification of pregnancies at risk for IUGR and the generation of clinical interventional strategies.
Collapse
|
46
|
Zhao H, Ozen M, Wong RJ, Stevenson DK. Heme oxygenase-1 in pregnancy and cancer: similarities in cellular invasion, cytoprotection, angiogenesis, and immunomodulation. Front Pharmacol 2015; 5:295. [PMID: 25642189 PMCID: PMC4294126 DOI: 10.3389/fphar.2014.00295] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/18/2014] [Indexed: 01/28/2023] Open
Abstract
Pregnancy can be defined as a “permissible” process, where a semi-allogeneic fetus and placenta are allowed to grow and survive within the mother. Similarly, in tumor growth, antigen-specific malignant cells proliferate and evade into normal tissues of the host. The microenvironments of the placenta and tumors are amazingly comparable, sharing similar mechanisms exploited by fetal or cancer cells with regard to surviving in a hypoxic microenvironment, invading tissues via degradation and vasculogenesis, and escaping host attack through immune privilege. Heme oxygease-1 (HO-1) is a stress-response protein that has antioxidative, anti-apoptotic, pro-angiogenic, and anti-inflammatory properties. Although a large volume of research has been published in recent years investigating the possible role(s) of HO-1 in pregnancy and in cancer development, the molecular mechanisms that regulate these “yin-yang” processes have still not been fully elucidated. Here, we summarize and compare pregnancy and cancer development, focusing primarily on the function of HO-1 in cellular invasion, cytoprotection, angiogenesis, and immunomodulation. Due to the similarities of both processes, a thorough understanding of the molecular mechanisms of each process may reveal and guide the development of new approaches to prevent not only pregnancy disorders; but also, to study cancer.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - Maide Ozen
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
47
|
Zenclussen ML, Linzke N, Schumacher A, Fest S, Meyer N, Casalis PA, Zenclussen AC. Heme oxygenase-1 is critically involved in placentation, spiral artery remodeling, and blood pressure regulation during murine pregnancy. Front Pharmacol 2015; 5:291. [PMID: 25628565 PMCID: PMC4292788 DOI: 10.3389/fphar.2014.00291] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/15/2014] [Indexed: 12/26/2022] Open
Abstract
The onset of pregnancy implies the appearance of a new organ, the placenta. One main function of the placenta is to supply oxygen to the fetus via hemoproteins. In this review, we highlight the importance of the enzyme heme oxygenase-1 (HO-1) for pregnancy to be established and maintained. HO-1 expression is pivotal to promote placental function and fetal development, thus determining the success of pregnancy. The deletion of the gene Hmox1 in mice leads to inadequate remodeling of spiral arteries and suboptimal placentation followed by intrauterine growth restriction (IUGR) and fetal lethality. A partial Hmox1 deletion leads to IUGR as well, with heterozygote and wild-type fetuses being born, but Hmox1 (-/-) significantly below the expected Mendelian rate. This strong phenotype is associated with diminished number of pregnancy-protective uterine natural killer (uNK) cells. Pregnant heterozygote females develop gestational hypertension. The protective HO-1 effects on placentation and fetal growth can be mimicked by the exogenous administration of carbon monoxide (CO), a product of heme catalyzed by HO-1. CO application promotes the in situ proliferation of uNK cells, restores placentation and fetal growth, while normalizing blood pressure. Similarly, HO-1 inhibition provokes hypertension in pregnant rats. The HO-1/CO axis plays a pivotal role in sustaining pregnancy and aids in the understanding of the biology of pregnancy and reveals a promising therapeutic application in the treatment of pregnancy complications.
Collapse
Affiliation(s)
- Maria L Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Nadja Linzke
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Stefan Fest
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Nicole Meyer
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Pablo A Casalis
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg , Germany
| |
Collapse
|
48
|
Schumacher A, Zenclussen AC. Effects of heme oxygenase-1 on innate and adaptive immune responses promoting pregnancy success and allograft tolerance. Front Pharmacol 2015; 5:288. [PMID: 25610397 PMCID: PMC4285018 DOI: 10.3389/fphar.2014.00288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/10/2014] [Indexed: 12/14/2022] Open
Abstract
The heme-degrading enzyme heme oxygenase-1 (HO-1) has cytoprotective, antioxidant, and anti-inflammatory properties. Moreover, HO-1 is reportedly involved in suppressing destructive immune responses associated with inflammation, autoimmune diseases, and allograft rejection. During pregnancy, maternal tolerance to foreign fetal antigens is a prerequisite for successful embryo implantation and fetal development. Here, HO-1 has been implicated in counteracting the overwhelming inflammatory immune responses towards fetal allo-antigens, thereby contributing to fetal acceptance. Accordingly, HO-1 ablation negatively impacts the critical steps of pregnancy such as fertilization, implantation, placentation, and fetal growth. In the present review, we summarize recent data on the immune modulatory capacity of HO-1 towards allo-antigens expressed by the semi-allogeneic fetus and organ allografts. In this regard, HO-1 has been shown to promote alloantigen tolerance by blocking dendritic cell maturation resulting in reduced T cell responses and increased numbers of regulatory T cells. Moreover, HO-1 is suggested to shift the uterine cytokine milieu towards a protective Th2 profile and protects fetal tissue from apoptosis by upregulating anti-apoptotic molecules. Thus, HO-1 is not only a pivotal regulator of the initial steps of pregnancy; but also, an important player in supporting the maternal immune system in tolerating the fetus.
Collapse
Affiliation(s)
- Anne Schumacher
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| | - Ana C Zenclussen
- Department of Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Germany
| |
Collapse
|
49
|
Iriyama T, Sun K, Parchim NF, Li J, Zhao C, Song A, Hart LA, Blackwell SC, Sibai BM, Chan LNL, Chan TS, Hicks MJ, Blackburn MR, Kellems RE, Xia Y. Elevated placental adenosine signaling contributes to the pathogenesis of preeclampsia. Circulation 2014; 131:730-41. [PMID: 25538227 DOI: 10.1161/circulationaha.114.013740] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Preeclampsia is a prevalent hypertensive disorder of pregnancy and a leading cause of maternal and neonatal morbidity and mortality worldwide. This pathogenic condition is speculated to be caused by placental abnormalities that contribute to the maternal syndrome. However, the specific factors and signaling pathways that lead to impaired placentas and maternal disease development remain elusive. METHODS AND RESULTS Using 2 independent animal models of preeclampsia (genetically engineered pregnant mice with elevated adenosine exclusively in placentas and a pathogenic autoantibody-induced preeclampsia mouse model), we demonstrated that chronically elevated placental adenosine was sufficient to induce hallmark features of preeclampsia, including hypertension, proteinuria, small fetuses, and impaired placental vasculature. Genetic and pharmacological approaches revealed that elevated placental adenosine coupled with excessive A₂B adenosine receptor (ADORA2B) signaling contributed to the development of these features of preeclampsia. Mechanistically, we provided both human and mouse evidence that elevated placental CD73 is a key enzyme causing increased placental adenosine, thereby contributing to preeclampsia. CONCLUSIONS We determined that elevated placental adenosine signaling is a previously unrecognized pathogenic factor for preeclampsia. Moreover, our findings revealed the molecular basis underlying the elevation of placental adenosine and the detrimental role of excess placental adenosine in the pathophysiology of preeclampsia, and thereby, we highlight novel therapeutic targets.
Collapse
Affiliation(s)
- Takayuki Iriyama
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Kaiqi Sun
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Nicholas F Parchim
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Jessica Li
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Cheng Zhao
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Anren Song
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Laura A Hart
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Sean C Blackwell
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Baha M Sibai
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Lee-Nien L Chan
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Teh-Sheng Chan
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - M John Hicks
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Michael R Blackburn
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Rodney E Kellems
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.)
| | - Yang Xia
- From Departments of Biochemistry and Molecular Biology (T.I., K.S., N.F.P., J.L., C.Z., A.S., M.R.B., R.E.K., Y.X.) and Department of Obstetrics, Gynecology, and Reproductive Sciences (L.A.H., S.C.B., B.M.S.), University of Texas Medical School at Houston: Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Japan (T.I.); Graduate School of Biomedical Sciences, University of Texas, Houston (K.S., N.F.P., M.R.B, R.E.K., Y.X.); Department of Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, (L-N. L.C., T.-S.C.); and Department of Pathology, Texas Children's Hospital, Houston (M.J.H.).
| |
Collapse
|
50
|
Goulopoulou S, Davidge ST. Molecular mechanisms of maternal vascular dysfunction in preeclampsia. Trends Mol Med 2014; 21:88-97. [PMID: 25541377 DOI: 10.1016/j.molmed.2014.11.009] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/11/2014] [Accepted: 11/26/2014] [Indexed: 12/13/2022]
Abstract
In preeclampsia, as a heterogeneous syndrome, multiple pathways have been proposed for both the causal as well as the perpetuating factors leading to maternal vascular dysfunction. Postulated mechanisms include imbalance in the bioavailability and activity of endothelium-derived contracting and relaxing factors and oxidative stress. Studies have shown that placenta-derived factors [antiangiogenic factors, microparticles (MPs), cell-free nucleic acids] are released into the maternal circulation and act on the vascular wall to modify the secretory capacity of endothelial cells and alter the responsiveness of vascular smooth muscle cells to constricting and relaxing stimuli. These molecules signal their deleterious effects on the maternal vascular wall via pathways that provide the molecular basis for novel and effective therapeutic interventions.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada.
| |
Collapse
|