1
|
Zhou C, Freel C, Mills O, Yang XR, Yan Q, Zheng J. MicroRNA-29 differentially mediates preeclampsia-dysregulated cellular responses to cytokines in female and male fetal endothelial cells. J Physiol 2023; 601:3631-3645. [PMID: 37401732 PMCID: PMC10807859 DOI: 10.1113/jp284746] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
Preeclampsia (PE) differentially impairs female and male fetal endothelial cell function, which is associated with an increased risk of adult-onset cardiovascular disorders in children born to mothers with PE. However, the underlying mechanisms are poorly defined. We hypothesize that dysregulation of microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in PE disturbs gene expression and cellular responses to cytokines in fetal endothelial cells in a fetal sex-dependent manner. RT-qPCR analysis of miR-29a/c-3p was performed on female and male unpassaged (P0) human umbilical vein endothelial cells (HUVECs) from normotensive (NT) pregnancies and PE. Bioinformatic analysis of an RNA-seq dataset was performed to identify PE-dysregulated miR-29a/c-3p target genes in female and male P0-HUVECs. Gain- and loss-of-function assays were conducted to determine the effects of miR-29a/c-3p on endothelial monolayer integrity and proliferation in response to transforming growth factor-β1 (TGFβ1) and tumour necrosis factor-α (TNFα) in NT and PE HUVECs at passage 1. We observed that PE downregulated miR-29a/c-3p in male and female P0-HUVECs. PE dysregulated significantly more miR-29a/c-3p target genes in female vs. male P0-HUVECs. Many of these PE-differentially dysregulated miR-29a/c-3p target genes are associated with critical cardiovascular diseases and endothelial function. We further demonstrated that miR-29a/c-3p knockdown specifically recovered the PE-abolished TGFβ1-induced strengthening of endothelial monolayer integrity in female HUVECs, while miR-29a/c-3p overexpression specifically enhanced the TNFα-promoted cell proliferation in male PE HUVECs. In conclusion, PE downregulates miR-29a/c-3p expression and differentially dysregulates miR-29a/c-3p target genes associated with cardiovascular diseases and endothelial function in female and male fetal endothelial cells, possibly contributing to the fetal sex-specific endothelial dysfunction observed in PE. KEY POINTS: Preeclampsia differentially impairs female and male fetal endothelial cell function in responses to cytokines. Pro-inflammatory cytokines are elevated in maternal circulation during pregnancy in preeclampsia. MicroRNAs are critical regulators of endothelial cell function during pregnancy. We have previously reported that preeclampsia downregulated microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in primary fetal endothelial cells. However, it is unknown if PE differentially dysregulates the expression of miR-29a/c-3p in female and male fetal endothelial cells. We show that preeclampsia downregulates miR-29a/c-3p in male and female HUVECs and preeclampsia dysregulates cardiovascular disease- and endothelial function-associated miR-29a/c-3p target genes in HUVECs in a fetal sex-specific manner. MiR-29a/c-3p differentially mediate cell responses to cytokines in female and male fetal endothelial cells from preeclampsia. We have revealed fetal sex-specific dysregulation of miR-29a/c-3p target genes in fetal endothelial cells from preeclampsia. This differential dysregulation may contribute to fetal sex-specific endothelial dysfunction in offspring born to preeclamptic mothers.
Collapse
Affiliation(s)
- Chi Zhou
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
- Department of Obstetrics and Gynecology, the University of Arizona, Tucson, AZ, United States
| | - Colman Freel
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Current Institution: University of Nebraska Medical Center, Omaha, NE, United States
| | - Olivia Mills
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Xin-Ran Yang
- School of Animal and Comparative Biomedical Sciences, the University of Arizona, Tucson, AZ, United States
| | - Qin Yan
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
2
|
Deer E, Herrock O, Campbell N, Cornelius D, Fitzgerald S, Amaral LM, LaMarca B. The role of immune cells and mediators in preeclampsia. Nat Rev Nephrol 2023; 19:257-270. [PMID: 36635411 PMCID: PMC10038936 DOI: 10.1038/s41581-022-00670-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 01/14/2023]
Abstract
Preeclampsia is a hypertensive disorder of major concern in pregnancy than can lead to intrauterine growth restriction, placental abruption and stillbirth. The pathophysiology of preeclampsia is multifactorial, including not only kidney dysfunction but also endothelial dysfunction, as the maternal endothelium becomes exposed to placental factors that are released into the circulation and increase systemic levels of vasoconstrictors, oxidative stress, anti-angiogenic factors and inflammatory mediators. Importantly, inflammation can lead to insufficient placental perfusion and low birthweight in offspring. Various innate and adaptive immune cells and mediators have been implicated in the development of preeclampsia, in which oxidative stress is associated with activation of the maternal inflammatory response. Immune cells such as regulatory T cells, macrophages, natural killer cells, and neutrophils are known to have major causative roles in the pathology of preeclampsia, but the contributions of additional immune cells such as B cells, inflammatory cytokines and anti-angiotensin II type 1 receptor autoantibodies are also now recognized. Immunological interventions, therefore, have therapeutic potential in this disease. Here, we provide an overview of the immune responses that are involved in the pathogenesis of preeclampsia, including the role of innate and adaptive immune cells and mediators.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Denise Cornelius
- Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sarah Fitzgerald
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
3
|
Zhou C, Freel C, Mills O, Yang XR, Yan Q, Zheng J. MicroRNA-29 Differentially Mediates Preeclampsia-Dysregulated Cellular Responses to Cytokines in Female and Male Fetal Endothelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.532827. [PMID: 36993536 PMCID: PMC10055181 DOI: 10.1101/2023.03.17.532827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Introduction Preeclampsia (PE) differentially impairs female and male fetal endothelial cell function which is associated with the increased risks of adult-onset cardiovascular disorders in children born to mothers with PE. However, the underlying mechanisms are poorly defined. We hypothesize that dysregulation of microRNA-29a-3p and 29c-3p (miR-29a/c-3p) in PE disturbs gene expression and cellular responses to cytokines in fetal endothelial cells in a fetal sex-dependent manner. Methods RT-qPCR analysis of miR-29a/c-3p was performed on female and male unpassaged (P0) human umbilical vein endothelial cells (HUVECs) from normotensive (NT) and PE pregnancies. Bioinformatic analysis of an RNAseq dataset was performed to identify PE-dysregulated miR-29a/c-3p target genes in female and male P0-HUVECs. Gain- and loss-of-function assays were conducted to determine the effects of miR-29a/c-3p on endothelial monolayer integrity and proliferation in response to TGFβ1 and TNFα in NT and PE HUVECs at passage 1. Results PE downregulated miR-29a/c-3p in male, but not female P0-HUVECs. PE dysregulated significantly more miR-29a/c-3p target genes in female vs. male P0-HUVECs. Many of these PE-differentially dysregulated miR-29a/c-3p target genes are associated with critical cardiovascular diseases and endothelial functions. We further demonstrated that miR-29a/c-3p knockdown specifically recovered the PE-abolished TGFβ1-induced strengthening of endothelial monolayer integrity in female HUVECs, while miR-29a/c-3p overexpression specifically enhanced the TNFα-promoted cell proliferation in male PE HUVECs. Conclusions PE differentially dysregulates miR-29a/c-3p and their target genes associated with cardiovascular diseases- and endothelial function in female and male fetal endothelial cells, possibly contributing to the fetal sex-specific endothelial dysfunction observed in PE.
Collapse
|
4
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Katoh Y, Iriyama T, Yano E, Sayama S, Seyama T, Kotajima-Murakami H, Sato A, Sakuma H, Iguchi Y, Yoshikawa M, Inaoka N, Ichinose M, Toshimitsu M, Sone K, Kumasawa K, Nagamatsu T, Ikeda K, Osuga Y. Increased production of inflammatory cytokines and activation of microglia in the fetal brain of preeclamptic mice induced by angiotensin II. J Reprod Immunol 2022; 154:103752. [PMID: 36202022 DOI: 10.1016/j.jri.2022.103752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/14/2022]
Abstract
Preeclampsia (PE) is a hypertensive obstetric disorder with poor prognosis for both the mother and offspring. Infants born to mothers with PE are known to be at increased risk of developing higher brain dysfunction, such as autism. However, how maternal PE can affect the environment in the fetal brain has not been fully elucidated. Here, we examined the impact of PE on the fetal brain in a mouse model of PE induced by angiotensin II (Ang II), focusing on changes in the inflammatory condition. We confirmed that pregnant mice which were continuously administered Ang II exhibited PE phenotypes, including high blood pressure, proteinuria, and fetal growth restriction. Quantitative RT-PCR analysis demonstrated that the brain of fetuses on embryonic day 17.5 (E17.5) in the Ang II-administered pregnant mice showed increased expression of cytokines, interleukin (IL)- 6, IL-17a, tumor necrosis factor-α, interferon-γ, IL-12, IL-4, and IL-10. Immunohistochemical analysis over a wide area, from the tip of the frontal lobe to the posterior cerebral end, on E17.5 revealed that the microglia in the fetal brain of the Ang II-administered group displayed higher solidity and circularity than those of the control group, indicating that the microglia had transformed to an amoeboid morphology and were activated. Our findings suggest that maternal PE may cause altered inflammatory conditions in the fetal brain, which might be associated with the pathological mechanism connecting maternal PE and brain dysfunction in the offspring.
Collapse
Affiliation(s)
- Yoshihisa Katoh
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan; Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan.
| | - Eriko Yano
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Seisuke Sayama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Takahiro Seyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Atsushi Sato
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
| | - Hiroshi Sakuma
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoshinobu Iguchi
- Technology Research Division, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Midori Yoshikawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Naoko Inaoka
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Mari Ichinose
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Masatake Toshimitsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Keiichi Kumasawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Zhai R, Liu Y, Tong J, Yu Y, Yang L, Gu Y, Niu J. Empagliflozin Ameliorates Preeclampsia and Reduces Postpartum Susceptibility to Adriamycin in a Mouse Model Induced by Angiotensin Receptor Agonistic Autoantibodies. Front Pharmacol 2022; 13:826792. [PMID: 35401209 PMCID: PMC8984158 DOI: 10.3389/fphar.2022.826792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/08/2022] [Indexed: 01/10/2023] Open
Abstract
Preeclampsia (PE) is the leading cause of maternal and perinatal morbidity and mortality and also is a risk factor for cardiovascular and kidney disease later in life. PE is associated with oversecretion of autoantibodies against angiotensin II type 1 receptor (AT1-AA) by the placenta into the maternal circulation. Here, we sought to determine the therapeutic value of the sodium-glucose co-transporter 2 (SGLT2) inhibitor empagliflozin (EMPA) in mice with AT1-AA-induced preeclampsia. Pregnant mice were injected with AT1-AA at gestation day (GD) 13 and treated daily with EMPA until GD 19, at which point some of the maternal mice were sacrificed and assessed. The other maternal mice were labored on time and challenged with adriamycin (ADR) at 12 weeks postpartum; their offspring were assessed for fetal outcomes. We showed that EMPA treatment significantly relieved high systolic blood pressure and proteinuria and ameliorated kidney injury in PE mice without affecting fetal outcomes. EMPA also ameliorated podocyte injury and oxidative stress, reduced the expression of SGLT2 and activated the AMPK/SIRT1 signaling pathway in vivo and in vitro. Remarkably, EMPA treatment during pregnancy reduced ADR-induced kidney and podocyte injury postpartum. These findings suggest that EMPA could be a potential pharmacological agent for PE.
Collapse
Affiliation(s)
- Ruonan Zhai
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yuan Liu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jiahao Tong
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Lin Yang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yong Gu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.,Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianying Niu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Yart L, Roset Bahmanyar E, Cohen M, Martinez de Tejada B. Role of the Uteroplacental Renin-Angiotensin System in Placental Development and Function, and Its Implication in the Preeclampsia Pathogenesis. Biomedicines 2021; 9:biomedicines9101332. [PMID: 34680449 PMCID: PMC8533592 DOI: 10.3390/biomedicines9101332] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022] Open
Abstract
Placental development and function implicate important morphological and physiological adaptations to thereby ensure efficient maternal–fetal exchanges, as well as pregnancy-specific hormone secretion and immune modulation. Incorrect placental development can lead to severe pregnancy disorders, such as preeclampsia (PE), which endangers both the mother and the infant. The implication of the systemic renin–angiotensin system (RAS) in the pregnancy-related physiological changes is now well established. However, despite the fact that the local uteroplacental RAS has been described for several decades, its role in placental development and function seems to have been underestimated. In this review, we provide an overview of the multiple roles of the uteroplacental RAS in several cellular processes of placental development, its implication in the regulation of placental function during pregnancy, and the consequences of its dysregulation in PE pathogenesis.
Collapse
Affiliation(s)
- Lucile Yart
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
| | | | - Marie Cohen
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
| | - Begoña Martinez de Tejada
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
- Correspondence:
| |
Collapse
|
8
|
Ye J, Que B, Huang Y, Lin Y, Chen J, Liu L, Shi Y, Wang Y, Wang M, Zeng T, Wang Z, Hu H, Xu Y, Shi L, Ye D, Liu J, Jiang H, Wan J, Ji Q. Interleukin-12p35 knockout promotes macrophage differentiation, aggravates vascular dysfunction, and elevates blood pressure in angiotensin II-infused mice. Cardiovasc Res 2020; 115:1102-1113. [PMID: 30395167 DOI: 10.1093/cvr/cvy263] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 09/06/2018] [Accepted: 10/31/2018] [Indexed: 12/11/2022] Open
Abstract
AIMS Numerous studies have demonstrated that inflammation is involved in the progression of hypertension. Inflammatory cytokines interleukin (IL)-12 and IL-35 belong to the IL-12 cytokine family and share the same IL-12p35 subunit. Accumulating evidence has demonstrated that IL-12p35 knockout (IL-12p35 KO) leads to cardiovascular disease by regulating the inflammatory response. This study aimed to investigate whether IL-12p35 KO elevates blood pressure in a hypertension mouse model. METHODS AND RESULTS Mice with angiotensin (Ang) II infusion showed marked aortic IL-12p35 expression; thus, aortic macrophages may be the main source of IL-12p35. Wild-type and IL-12p35 KO mice were infused with Ang II or saline. IL-12p35 KO promoted M1 macrophage differentiation, amplified the inflammatory response, aggravated vascular dysfunction, and elevated blood pressure in Ang II-treated mice. Then, some Ang II-infused mice were given phosphate buffer saline, mouse recombinant IL-12 (rIL-12), or rIL-35, and the results showed that rIL-12 but not rIL-35 treatment had an antihypertensive effect on Ang II-infused mice. In addition, detection of human plasma IL-12 levels in hypertensive patients and control subjects showed that IL-12 was significantly increased in hypertensive patients when compared with control subjects. In hypertensive patients, IL-12 levels were positively correlated with blood pressure. CONCLUSION IL-12p35 KO amplifies the inflammatory response and promotes blood pressure elevation in Ang II-treated mice. In addition, IL-12, but not IL-35, plays a protective role in the Ang II-induced hypertension model. Thus, IL-12 may be a novel therapeutic agent for the prevention and treatment of clinical hypertension.
Collapse
Affiliation(s)
- Jing Ye
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China.,Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Que
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Ying Huang
- Department of Ultrasound, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jiangbin Chen
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Tao Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Haiying Hu
- Department of Cardiology, Handan First Hospital, Handan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lei Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingwei Ji
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China.,Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
9
|
Gatford KL, Andraweera PH, Roberts CT, Care AS. Animal Models of Preeclampsia: Causes, Consequences, and Interventions. Hypertension 2020; 75:1363-1381. [PMID: 32248704 DOI: 10.1161/hypertensionaha.119.14598] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preeclampsia is a common pregnancy complication, affecting 2% to 8% of pregnancies worldwide, and is an important cause of both maternal and fetal morbidity and mortality. Importantly, although aspirin and calcium are able to prevent preeclampsia in some women, there is no cure apart from delivery of the placenta and fetus, often necessitating iatrogenic preterm birth. Preclinical models of preeclampsia are widely used to investigate the causes and consequences of preeclampsia and to evaluate safety and efficacy of potential preventative and therapeutic interventions. In this review, we provide a summary of the published preclinical models of preeclampsia that meet human diagnostic criteria, including the development of maternal hypertension, together with new-onset proteinuria, maternal organ dysfunction, and uteroplacental dysfunction. We then discuss evidence from preclinical models for multiple causal factors of preeclampsia, including those implicated in early-onset and late-onset preeclampsia. Next, we discuss the impact of exposure to a preeclampsia-like environment for later maternal and progeny health. The presence of long-term impairment, particularly cardiovascular outcomes, in mothers and progeny after an experimentally induced preeclampsia-like pregnancy, implies that later onset or reduced severity of preeclampsia will improve later maternal and progeny health. Finally, we summarize published intervention studies in preclinical models and identify gaps in knowledge that we consider should be targets for future research.
Collapse
Affiliation(s)
- Kathryn L Gatford
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Prabha H Andraweera
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Claire T Roberts
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Alison S Care
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| |
Collapse
|
10
|
Martell Claros N, Asenjo de la Fuente JE, Abad Cardiel M, García Donaire JA, Herráiz MA. [Role of the renin-angiotensin system in pregnancy and preeclampsia]. HIPERTENSION Y RIESGO VASCULAR 2020; 37:72-77. [PMID: 32147515 DOI: 10.1016/j.hipert.2020.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/04/2019] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
Abstract
The renin-angiotensin system (ARS) is a hormonal cascade that regulates blood pressure, electrolytes and water balance. AngiotensinII (AII) exerts its effects through the AT1 and AT2 receptors. AT1 is found in the syncytiotrophoblast, AT2 predominates during foetal development and its stimulation inhibits cell growth, increases apoptosis, causes vasodilation and regulates the development of foetal tissue. There is also an SRA in the placenta. The local generation of AII is responsible for the activation of AT1 receptors in the trophoblast. In normal pregnancy, concomitantly with reduction of blood pressure the circulating RAS increases, but blood pressure does not rise due to AII refractoriness, which does not occur in preeclampsia. We review the role of the SRA in normal pregnancy and preeclampsia.
Collapse
Affiliation(s)
- N Martell Claros
- Unidad de Hipertensión y Riesgo Vascular, Servicio de Medicina Interna, Hospital Clínico San Carlos, Madrid, España; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, España; Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, España.
| | - J E Asenjo de la Fuente
- Unidad de Ecografía y Diagnóstico Prenatal, Instituto de Salud de la Mujer JBLL, Hospital Clínico San Carlos, Madrid, España
| | - M Abad Cardiel
- Unidad de Hipertensión y Riesgo Vascular, Servicio de Medicina Interna, Hospital Clínico San Carlos, Madrid, España; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, España; Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, España
| | - J A García Donaire
- Unidad de Hipertensión y Riesgo Vascular, Servicio de Medicina Interna, Hospital Clínico San Carlos, Madrid, España; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, España
| | - M A Herráiz
- Instituto de Salud de la Mujer JBLL, Hospital Clínico San Carlos, Madrid, España
| |
Collapse
|
11
|
Vaka VR, Cunningham MW, Deer E, Franks M, Ibrahim T, Amaral LM, Usry N, Cornelius DC, Dechend R, Wallukat G, LaMarca BD. Blockade of endogenous angiotensin II type I receptor agonistic autoantibody activity improves mitochondrial reactive oxygen species and hypertension in a rat model of preeclampsia. Am J Physiol Regul Integr Comp Physiol 2019; 318:R256-R262. [PMID: 31721604 DOI: 10.1152/ajpregu.00179.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Preeclampsia (PE) is characterized by new-onset hypertension that usually occurs in the third trimester of pregnancy and is associated with oxidative stress and angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs). Inhibition of the AT1-AAs in the reduced uterine perfusion pressure (RUPP) rat, a model of PE, attenuates hypertension and many other characteristics of PE. We have previously shown that mitochondrial oxidative stress (mtROS) is a newly described PE characteristic exhibited by the RUPP rat that contributes to hypertension. However, the factors that cause mtROS in PE or RUPP are unknown. Thus, the objective of the current study is to use pharmacologic inhibition of AT1-AAs to examine their role in mtROS in the RUPP rat model of PE. AT1-AA inhibition in RUPP rats was achieved by administration of an epitope-binding peptide ('n7AAc'). Female Sprague-Dawley rats were divided into the following two groups: RUPP and RUPP + AT1-AA inhibition (RUPP + 'n7AAc'). On day 14 of gestation (GD), RUPP surgery was performed; 'n7AAc' peptide (2 µg/μL) was administered by miniosmotic pumps in a subset of RUPP rats; and on GD19, sera, placentas, and kidneys were collected. mitochondrial respiration and mtROS were measured in isolated mitochondria using the Oxygraph 2K and fluorescent microplate reader, respectively. Placental and renal mitochondrial respiration and mtROS were improved in RUPP + 'n7AAc' rats compared with RUPP controls. Moreover, endothelial cells (human umbilical vein endothelial cells) treated with RUPP + 'n7AAc' sera exhibited less mtROS compared with those treated with RUPP sera. Overall, our findings suggest that AT1-AA signaling is one stimulus of mtROS during PE.
Collapse
Affiliation(s)
- Venkata Ramana Vaka
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mark W Cunningham
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael Franks
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nathan Usry
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Berlin, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center, Charité Campus Buch, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Babbette D LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Obstetrics & Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
12
|
Encinas JFA, Foncesca CH, Perez MM, Simões DP, da Costa Aguiar Alves B, Bacci MR, Maifrino LBM, Fonseca FLA, da Veiga GL. Role of hypoxia-inducible factor 1α as a potential biomarker for renal diseases-A systematic review. Cell Biochem Funct 2019; 37:443-451. [PMID: 31317578 DOI: 10.1002/cbf.3425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/01/2019] [Accepted: 06/26/2019] [Indexed: 01/14/2023]
Abstract
Renal cells need oxygen for homeostasis; it is known for adjusting cellular functioning and the energy obtainment have a broad relationship with cellular respiration, through the O2 bioavailability. O2 homeostasis regulation in the kidney is mediated by hypoxia-inducible factors (HIFs). HIF is divided into three α isoforms, represented by HIF-1α, HIF-2α, and HIF-3α in addition to three paralogs of HIF-1β; these are involved in some metabolic processes, as well as in the pathogenesis of several diseases. Renal biopsy analyses of patients and experimental animal models aim to understand the relationship between HIF and protection against developing renal diseases or the induction of their onset, being thus this molecule can be considered a potential biomarker of renal disease. We carried out a systematic review to which we included studies on HIF-1α and renal disease in the last 5 years (2013-2018) in researches with humans and/or animal model through searches in three databases: LILACS, PubMed, and SciELO by two researchers. We obtained 22 articles that discussed the relationship with HIF as inductor or protector against renal disease and no relation between HIF and renal. We observed controversies remain regarding the relation between of HIF with renal diseases; this may be related to the different intracellular pathways mediated by HIF-1α, thereby determining differentiated cellular responses.
Collapse
Affiliation(s)
| | - Carlos Henrique Foncesca
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil
| | - Matheus Moreira Perez
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil
| | - Diogo Pimenta Simões
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil.,Universidade Municipal de São Caetano do Sul/USCS - São Caetano do Sul, Sao Caetano do Sul, Brazil
| | | | - Marcelo Rodrigues Bacci
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil
| | | | - Fernando Luiz Affonso Fonseca
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil.,Universidade Federal de São Paulo/UNIFESP - Diadema, São Paulo, Brazil
| | - Glaucia Luciano da Veiga
- Department of Clinical Analysis, Faculdade de Medicina do ABC/FMABC - Santo André, Santo André, Brazil
| |
Collapse
|
13
|
[Potential value of placental angiogenic factors as biomarkers in preeclampsia for clinical physicians]. Nephrol Ther 2019; 15:413-429. [PMID: 30935786 DOI: 10.1016/j.nephro.2018.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/10/2018] [Accepted: 10/14/2018] [Indexed: 12/20/2022]
Abstract
The role of angiogenic factors in the onset of clinical manifestations of preeclampsia was demonstrated in 2003 by the implication of sFlt-1, PlGF and VEGF, and in 2006 by the implication of soluble endoglin. Placental ischemia and inflammation observed in preeclampsia alter both the production and progression of angiogenic factors during pregnancy. During the first trimester, the combination of PlGF with clinical, biophysical and biological factors results in a better test than the conventional one. However, the clinical value of this method remains to be confirmed. During the second and third trimesters, the sFlt-1/PlGF ratio may be used, with or without pre-existing renal disease, for short-term prediction, diagnosis, and prognosis, and to evaluate the effectiveness of preeclampsia treatment. While a sFlt-1/PlGF ratio<38 and≤33, respectively, rules out the short-term onset and diagnosis of preeclampsia, a sFlt-1/PlGF ratio≥85 between 20 and 34 weeks of pregnancy and≥110 beyond 34 weeks of pregnancy confirms a diagnosis of preeclampsia. Angiogenic and non-angiogenic preeclampsia are identified by a sFlt-1PlGF≥85 and<85, respectively, with the risk of maternal and fetal complications at two weeks differing between the two. Similarly, a sFlt-1/PlGF ratio>665 and>205, respectively, is a good short-term predictor of adverse outcomes of early and late-onset preeclampsia. These values could be incorporated into future guidelines for better clinical management of preeclampsia.
Collapse
|
14
|
Zhang S, Wei M, Yue M, Wang P, Yin X, Wang L, Yang X, Liu H. Hyperinsulinemia precedes insulin resistance in offspring rats exposed to angiotensin II type 1 autoantibody in utero. Endocrine 2018; 62:588-601. [PMID: 30101377 DOI: 10.1007/s12020-018-1700-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/26/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Insulin resistance is highly associated with an adverse intrauterine environment. We previously reported that fetal rats exposed to angiotensin II type 1 receptor (AT1R) autoantibody (AT1-AA) displayed increased susceptibility to metabolic diseases during middle age. However, the timing of the onset of insulin resistance remains unknown. In this study, we examined the offspring of AT1-AA-positive rats, tracking the development of insulin resistance. METHODS Pregnant rats were intravenously injected with AT1-AA. Afterwards, we collected serum samples and liver tissues of the offspring at various stages, including gestation day 18, 3 weeks (weaning period), 18 weeks (young adulthood), and 48 weeks (middle age) after birth. RESULTS Compared with saline control group, hepatic vacuolar degeneration was visible in AT1-AA offspring rats as early as 3 weeks; hyperinsulinemia and impaired glucose tolerance occurred at 18 weeks of age, however, insulin resistance was not observed until 48 weeks. At 18 weeks we detected suppressed protein levels of insulin receptor (IR) but increased levels of IR substrate 1 (IRS1) in the liver of AT1-AA group rats. Interestingly, both IR and IRS1/2 were significantly decreased at 48 weeks. Liver proteomic analysis indicated that the differences in protein expression between the AT1-AA and control rats became more pronounced with age, particularly in terms of mitochondrial energy metabolism. CONCLUSION Rats exposed to AT1-AA in utero developed hyperinsulinemia from young adulthood which subsequently progressed to insulin resistance, and was linked with abnormal hepatic structure and impaired IR signaling. Additionally, dysregulation of energy metabolism may play a fundamental role in predisposing offspring to insulin resistance.
Collapse
Affiliation(s)
- Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingming Wei
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingming Yue
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Pengli Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaochen Yin
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoli Yang
- Department of Reproductive Center, Taiyuan Central Hospital, Taiyuan, Shanxi, China
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Tayyar AT, Tayyar A, Kozali S, Karakus R, Eser A, Abide Yayla C, Yalcin ET, Dag I, Eroglu M. Maternal serum sestrin 2 levels in preeclampsia and their relationship with the severity of the disease. Hypertens Pregnancy 2018; 38:13-19. [DOI: 10.1080/10641955.2018.1540702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Ahter Tanay Tayyar
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Acıbadem University Faculty of Medicine, İstanbul, Turkey
| | - Ahmet Tayyar
- Department of Obstetrics and Gynecology, Medipol University Faculty of Medicine, Istanbul, Turkey
| | - Sukran Kozali
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| | - Resul Karakus
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| | - Ahmet Eser
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| | - Cigdem Abide Yayla
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| | - Emel Tugce Yalcin
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| | - Ismail Dag
- Department of Clinical Biochemistry, Eyup State Hospital, Istanbul, Turkey
| | - Mustafa Eroglu
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Maternity and Children’s Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
16
|
Small HY, Migliarino S, Czesnikiewicz-Guzik M, Guzik TJ. Hypertension: Focus on autoimmunity and oxidative stress. Free Radic Biol Med 2018; 125:104-115. [PMID: 29857140 DOI: 10.1016/j.freeradbiomed.2018.05.085] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022]
Abstract
Understanding the causal role of the immune and inflammatory responses in hypertension has led to questions regarding the links between hypertension and autoimmunity. Immune pathology in primary hypertension mimics several autoimmune mechanisms observed in the pathogenesis of systemic lupus erythematosus, psoriasis, systemic sclerosis, rheumatoid arthritis and periodontitis. More importantly, the prevalence of hypertension in patients with these autoimmune diseases is significantly increased, when compared to control populations. Clinical and epidemiological evidence is reviewed along with possible mechanisms linking hypertension and autoimmunity. Inflammation and oxidative stress are linked in a self-perpetuating cycle that significantly contributes to the vascular dysfunction and renal damage associated with hypertension. T cell, B cell, macrophage and NK cell infiltration into these organs is essential for this pathology. Effector cytokines such as IFN-γ, TNF-α and IL-17 affect Na+/H+ exchangers in the kidney. In blood vessels, they lead to endothelial dysfunction and loss of nitric oxide bioavailability and cause vasoconstriction. Both renal and vascular effects are, in part, mediated through induction of reactive oxygen species-producing enzymes such as superoxide anion generating NADPH oxidases and dysfunction of anti-oxidant systems. These mechanisms have recently become important therapeutic targets of novel therapies focused on scavenging oxidative (isolevuglandin) modification of neo-antigenic peptides. Effects of classical immune targeted therapies focused on immunosuppression and anti-cytokine treatments are also reviewed.
Collapse
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Serena Migliarino
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Dental Prophylaxis and Experimental Dentistry, Dental School of Jagiellonian University, Krakow, Poland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.
| |
Collapse
|
17
|
Armaly Z, Jadaon JE, Jabbour A, Abassi ZA. Preeclampsia: Novel Mechanisms and Potential Therapeutic Approaches. Front Physiol 2018; 9:973. [PMID: 30090069 PMCID: PMC6068263 DOI: 10.3389/fphys.2018.00973] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023] Open
Abstract
Preeclampsia is a serious complication of pregnancy where it affects 5–8% of all pregnancies. It increases the morbidity and mortality of both the fetus and pregnant woman, especially in developing countries. It deleteriously affects several vital organs, including the kidneys, liver, brain, and lung. Although, the pathogenesis of preeclampsia has not yet been fully understood, growing evidence suggests that aberrations in the angiogenic factors levels and coagulopathy are responsible for the clinical manifestations of the disease. The common nominator of tissue damage of all these target organs is endothelial injury, which impedes their normal function. At the renal level, glomerular endothelial injury leads to the development of maternal proteinuria. Actually, peripheral vasoconstriction secondary to maternal systemic inflammation and endothelial cell activation is sufficient for the development of preeclampsia-induced hypertension. Similarly, preeclampsia can cause hepatic and neurologic dysfunction due to vascular damage and/or hypertension. Obviously, preeclampsia adversely affects various organs, however it is not yet clear whether pre-eclampsia per se adversely affects various organs or whether it exposes underlying genetic predispositions to cardiovascular disease that manifest in later life. The current review summarizes recent development in the pathogenesis of preeclampsia with special focus on novel diagnostic biomarkers and their relevance to potential therapeutic options for this disease state. Specifically, the review highlights the renal manifestations of the disease with emphasis on the involvement of angiogenic factors in vascular injury and on how restoration of the angiogenic balance affects renal and cardiovascular outcome of Preeclamptic women.
Collapse
Affiliation(s)
- Zaher Armaly
- Department of Nephrology, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Jimmy E Jadaon
- Department of Obstetrics and Gynecology, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel.,Laboratory Medicine, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Adel Jabbour
- Laboratory Medicine, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Zaid A Abassi
- Department of Physiology, The Ruth and Burce Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel.,Department of Laboratory Medicine, Rambam Health Campus, Haifa, Israel
| |
Collapse
|
18
|
Marshall SA, Hannan NJ, Jelinic M, Nguyen TP, Girling JE, Parry LJ. Animal models of preeclampsia: translational failings and why. Am J Physiol Regul Integr Comp Physiol 2018; 314:R499-R508. [DOI: 10.1152/ajpregu.00355.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia affects up to 8% of pregnancies worldwide and is a leading cause of both maternal and fetal morbidity and mortality. Our current understanding of the cause(s) of preeclampsia is far from complete, and the lack of a single reliable animal model that recapitulates all aspects of the disease further confounds our understanding. This is partially due to the heterogeneous nature of the disease, coupled with our evolving understanding of its etiology. Nevertheless, animal models are still highly relevant and useful tools that help us better understand the pathophysiology of specific aspects of preeclampsia. This review summarizes the various types and characteristics of animal models used to study preeclampsia, highlighting particular features of these models relevant to clinical translation. This review points out the strengths and limitations of these models to illustrate the importance of using the appropriate model depending on the research question.
Collapse
Affiliation(s)
- Sarah A. Marshall
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Natalie J. Hannan
- The Translational Obstetrics Group, Mercy Hospital for Women, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
| | - Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Thy P.H. Nguyen
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E. Girling
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, The University of Melbourne and Royal Women’s Hospital, Parkville, Victoria, Australia
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Ye J, Ji Q, Liu J, Liu L, Huang Y, Shi Y, Shi L, Wang M, Liu M, Feng Y, Jiang H, Xu Y, Wang Z, Song J, Lin Y, Wan J. Interleukin 22 Promotes Blood Pressure Elevation and Endothelial Dysfunction in Angiotensin II-Treated Mice. J Am Heart Assoc 2017; 6:e005875. [PMID: 28974499 PMCID: PMC5721831 DOI: 10.1161/jaha.117.005875] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND CD4+ T helper (Th) cells, including Th1, Th2, and Th17 cells, play critical roles in angiotensin II-induced hypertension. Th22 cells, a novel subset of Th cells, take part in cardiovascular diseases by producing IL-22 (interleukin 22). This study aimed to investigate whether IL-22 is involved in hypertension. METHODS AND RESULTS Th22 cells and IL-22 levels were detected in angiotensin II-infused mice, and the results showed that Th22 cells and IL-22 levels significantly increased. To determine the effect of Th22/IL-22 on blood pressure regulation, angiotensin II-infused mice were treated with recombinant mouse IL-22, an anti-IL-22 neutralizing monoclonal antibody, or control. Treatment with recombinant IL-22 resulted in increased blood pressure, amplified inflammatory responses, and aggravated endothelial dysfunction, whereas the anti-IL-22 neutralizing monoclonal antibody decreased blood pressure, reduced inflammatory responses, and attenuated endothelial dysfunction. To determine whether the STAT3 (signal transducer and activator of transcription 3) pathway mediates the effect of IL-22 on blood pressure regulation, the special STAT3 pathway inhibitor S31-201 was administered to mice treated with recombinant IL-22. S31-201 treatment significantly ameliorated the IL-22 effects of increased blood pressure and endothelial dysfunction. In addition, serum IL-22 levels were significantly increased in hypertensive patients compared with healthy persons. Correlation analysis showed a positive correlation between IL-22 levels and blood pressure. CONCLUSIONS IL-22 amplifies the inflammatory response, induces endothelial dysfunction and promotes blood pressure elevation in angiotensin II-induced hypertensive mice. The STAT3 pathway mediates the effect of IL-22 on hypertension. Blocking IL-22 may be a novel therapeutic strategy to prevent and treat hypertension.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengling Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ying Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junlong Song
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
20
|
Lopez Gelston CA, Mitchell BM. Recent Advances in Immunity and Hypertension. Am J Hypertens 2017; 30:643-652. [PMID: 28200062 DOI: 10.1093/ajh/hpx011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/18/2017] [Indexed: 01/01/2023] Open
Abstract
Persistent immune system activation plays an important role in the development of various forms of hypertension. Activation of the innate immune system, inflammation, and subsequent adaptive immune system response causing end-organ injury and dysfunction ultimately leads to hypertension and its associated sequelae including coronary artery disease, heart failure, stroke, and chronic kidney disease. In this review, we will provide updates on the innate and adaptive immune cells involved in hypertension, the current understanding of how the immune system gets activated, and examine the recently discovered mechanisms involved in several forms of experimental hypertension.
Collapse
Affiliation(s)
- Catalina A Lopez Gelston
- Department of Medical Physiology, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University Health Science Center, College Station, Texas, USA
| |
Collapse
|
21
|
Aggarwal S, Sunderland N, Thornton C, Xu B, Hennessy A, Makris A. A longitudinal analysis of angiotensin II type 1 receptor antibody and angiogenic markers in pregnancy. Am J Obstet Gynecol 2017; 216:170.e1-170.e8. [PMID: 27793555 DOI: 10.1016/j.ajog.2016.10.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/28/2016] [Accepted: 10/18/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Preeclampsia can be caused by shallow trophoblast invasion and results in endothelial dysfunction. Angiotensin II type 1 receptor antibodies may have a role in both processes. Other angiogenic markers (placental growth factor, soluble fms-like tyrosine kinase-1, and soluble endoglin) have been shown to alter before clinically evident preeclampsia. OBJECTIVE The aim of this study is to assess the longitudinal changes and utility of biomarker angiotensin II type 1 receptor antibodies and angiogenic markers in hypertensive disorders of pregnancy, gestational hypertension, and preeclampsia. STUDY DESIGN A longitudinal prospective cohort observational study of angiogenic markers and a secondary retrospective case-control study of angiotensin II type 1 receptor antibody changes were conducted. The studies were conducted in a large tertiary metropolitan teaching hospital (Sydney, Australia). Sequential recruitment of women with a singleton pregnancy (N = 351) was undertaken. Plasma concentrations of angiotensin II type 1 receptor antibodies, placental growth factor, soluble fms-like tyrosine kinase-1, and soluble endoglin were measured using validated enzyme-linked immunosorbent assays at 12, 18, 28, 36, and 40 weeks' gestation and 6 weeks' postpartum. Clinical, demographic, and pregnancy data were prospectively collected. Pregnancy outcomes were classified as normotensive, gestational hypertension, or preeclampsia. Analyses were carried out using software and significance set at P < .05. RESULTS In all, 351 women were recruited, 17 developed gestational hypertension, and 18 developed preeclampsia. Women with preeclampsia at baseline were heavier (P = .015), were taller (P = .046), and had higher systolic (P = .029) and diastolic (P = .006) blood pressure. The preeclampsia group had higher soluble fms-like tyrosine kinase-1 from ≥28 weeks (P = .003) and lower placental growth factor from 18 weeks (P = .004). Soluble endoglin and angiotensin II type 1 receptor antibodies did not vary over time or between groups. Angiotensin II type 1 receptor antibody (12 weeks) was positively correlated with serum pregnancy associated plasma protein A (P = .008) and human chorionic gonadotrophin (P = .04). CONCLUSION Angiogenic markers vary longitudinally during pregnancy and placental growth factor and soluble fms-like tyrosine kinase-1 have a role for predicting and diagnosing preeclampsia later in disease. Our data show that angiotensin II type 1 receptor antibodies are not sensitive for disease and hence not useful as a biomarker. Larger studies are required to describe the role and functionality of angiotensin II type 1 receptor antibodies in preeclampsia.
Collapse
Affiliation(s)
- Shikha Aggarwal
- School of Medicine, Western Sydney University, Sydney, Australia; Heart Research Institute, Sydney, Australia.
| | | | - Charlene Thornton
- Department of Renal Medicine, South Western Sydney Local Health District, Sydney, Australia
| | - Bei Xu
- Heart Research Institute, Sydney, Australia
| | - Annemarie Hennessy
- School of Medicine, Western Sydney University, Sydney, Australia; Heart Research Institute, Sydney, Australia; Department of Renal Medicine, South Western Sydney Local Health District, Sydney, Australia
| | - Angela Makris
- School of Medicine, Western Sydney University, Sydney, Australia; Heart Research Institute, Sydney, Australia; Department of Renal Medicine, South Western Sydney Local Health District, Sydney, Australia
| |
Collapse
|
22
|
Luo R, Liu C, Elliott SE, Wang W, Parchim N, Iriyama T, Daugherty PS, Tao L, Eltzschig HK, Blackwell SC, Sibai BM, Kellems RE, Xia Y. Transglutaminase is a Critical Link Between Inflammation and Hypertension. J Am Heart Assoc 2016; 5:JAHA.116.003730. [PMID: 27364991 PMCID: PMC5015405 DOI: 10.1161/jaha.116.003730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background The pathogenesis of essential hypertension is multifactorial with different underlying mechanisms contributing to disease. We have recently shown that TNF superfamily member 14 LIGHT (an acronym for homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, also known as TNFSF14) induces hypertension when injected into mice. Research reported here was undertaken to examine the role of transglutaminase (TGase) in LIGHT‐induced hypertension. Methods and Results Initial experiments showed that plasma and kidney TGase activity was induced by LIGHT infusion (13.91±2.92 versus 6.75±1.92 mU/mL and 19.86±3.55 versus 12.00±0.97 mU/10 μg) and was accompanied with hypertension (169±7.16 versus 117.17±11.57 mm Hg at day 14) and renal impairment (proteinuria, 61.33±23.21 versus 20.38±9.01 μg/mg; osmolality, 879.57±93.02 versus 1407.2±308.04 mmol/kg). The increase in renal TGase activity corresponded to an increase in RNA for the tissue TGase isoform, termed TG2. Pharmacologically, we showed that LIGHT‐induced hypertension and renal impairment did not occur in the presence of cystamine, a well‐known competitive inhibitor of TGase activity. Genetically, we showed that LIGHT‐mediated induction of TGase, along with hypertension and renal impairment, was dependent on interleukin‐6 and endothelial hypoxia inducible factor‐1α. We also demonstrated that interleukin‐6, endothelial hypoxia inducible factor‐1α, and TGase are required for LIGHT‐induced production of angiotensin receptor agonistic autoantibodies. Conclusions Thus, LIGHT‐induced hypertension, renal impairment, and production of angiotensin receptor agonistic autoantibodies require TGase, most likely the TG2 isoform. Our findings establish TGase as a critical link between inflammation, hypertension, and autoimmunity.
Collapse
Affiliation(s)
- Renna Luo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chen Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX
| | - Serra E Elliott
- Department of Chemical Engineering, University of California, Santa Barbara, CA
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Nicholas Parchim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX
| | - Takayuki Iriyama
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Obstetrics and Gynecology, University of Tokyo, Japan
| | - Patrick S Daugherty
- Department of Chemical Engineering, University of California, Santa Barbara, CA
| | - Lijian Tao
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Holger K Eltzschig
- Department of Anesthesiology, University of Colorado-Medical School, Denver, CO
| | - Sean C Blackwell
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Texas Health Science Center at Houston, TX
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Texas Health Science Center at Houston, TX
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX The University of Texas Graduate School of Biomedical Sciences at Houston, TX
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX The University of Texas Graduate School of Biomedical Sciences at Houston, TX
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Aggarwal S, Makris A, Hennessy A. Linking the old and new -- do angiotensin II type 1 receptor antibodies provide the missing link in the pathophysiology of preeclampsia? Hypertens Pregnancy 2015; 34:369-82. [PMID: 26153629 DOI: 10.3109/10641955.2015.1051227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Preeclampsia remains a leading cause of maternal and neonatal morbidity and mortality. The pathophysiology of preeclampsia remains poorly understood with various pathological mechanisms being implicated including the renin angiotensin system (RAAS), angiogenic pathways and various components of the immune system. Recently a pathogenic autoimmune factor has been identified in the form of auto-agonistic angiotensin II type 1 receptor antibodies (AT1-AA). AT1-AA have been studied in vitro and in vivo in various human and animal models and these data have provided compelling evidence for their role in preeclampsia. This review summarises the current literature surrounding the role of AT1-AA in preeclampsia and draws links between this relatively novel antibody to well-established pathological mechanisms including the immune system, the RAAS, angiogenic pathways and placental ischaemia.
Collapse
Affiliation(s)
- Shikha Aggarwal
- School of Medicine, University of Western Sydney , NSW , Australia
| | | | | |
Collapse
|
26
|
Luo R, Zhang W, Zhao C, Zhang Y, Wu H, Jin J, Zhang W, Grenz A, Eltzschig HK, Tao L, Kellems RE, Xia Y. Elevated Endothelial Hypoxia-Inducible Factor-1α Contributes to Glomerular Injury and Promotes Hypertensive Chronic Kidney Disease. Hypertension 2015; 66:75-84. [PMID: 25987665 DOI: 10.1161/hypertensionaha.115.05578] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 12/28/2022]
Abstract
Hypertensive chronic kidney disease is one of the most prevalent medical conditions with high morbidity and mortality in the United States and worldwide. However, early events initiating the progression to hypertensive chronic kidney disease are poorly understood. We hypothesized that elevated endothelial hypoxia-inducible factor-1α (HIF-1α) is a common early insult triggering initial glomerular injury leading to hypertensive chronic kidney disease. To test our hypothesis, we used an angiotensin II infusion model of hypertensive chronic kidney disease to determine the specific cell type and mechanisms responsible for elevation of HIF-1α and its role in the progression of hypertensive chronic kidney disease. Genetic studies coupled with reverse transcription polymerase chain reaction profiling revealed that elevated endothelial HIF-1α is essential to initiate glomerular injury and progression to renal fibrosis by the transcriptional activation of genes encoding multiple vasoactive proteins. Mechanistically, we found that endothelial HIF-1α gene expression was induced by angiotensin II in a nuclear factor-κB-dependent manner. Finally, we discovered reciprocal positive transcriptional regulation of endothelial Hif-1α and Nf-κb genes is a key driving force for their persistent activation and disease progression. Overall, our findings revealed that the stimulation of HIF-1α gene expression in endothelial cells is detrimental to induce kidney injury, hypertension, and disease progression. Our findings highlight early diagnostic opportunities and therapeutic approaches for hypertensive chronic kidney disease.
Collapse
Affiliation(s)
- Renna Luo
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Weiru Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Cheng Zhao
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Yujin Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Hongyu Wu
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Jianping Jin
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Wenzheng Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Almut Grenz
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Holger K Eltzschig
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Lijian Tao
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Rodney E Kellems
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Yang Xia
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.).
| |
Collapse
|
27
|
Bounds KR, Newell-Rogers MK, Mitchell BM. Four Pathways Involving Innate Immunity in the Pathogenesis of Preeclampsia. Front Cardiovasc Med 2015; 2:20. [PMID: 26664892 PMCID: PMC4671354 DOI: 10.3389/fcvm.2015.00020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/14/2015] [Indexed: 11/13/2022] Open
Abstract
The maternal innate immune system plays an important role both in normal pregnancy as well as hypertensive disorders of pregnancy including preeclampsia (PE). We propose four pathways that involve excessive innate immunity that lead to most forms of PE. Pre-existing endothelial dysfunction plus pregnancy leads to an excessive innate immune response resulting in widespread inflammation, placental and renal dysfunction, vasoconstriction, and PE. Placental dysfunction due to shallow trophoblast invasion, inadequate spiral artery remodeling, and/or low placental perfusion initiates an innate immune response leading to excessive inflammation, endothelial and renal dysfunction, and PE. A heightened innate immune system due to pre-existing or acquired infections plus the presence of a paternally derived placenta and semi-allogeneic fetus cause an excessive innate immune response which manifests as PE. Lastly, an abnormal and excessive maternal immune response to pregnancy leads to widespread inflammation, organ dysfunction, and PE. We discuss the potential role of innate immunity in each of these scenarios, as well as the overlap, and how targeting the innate immune system might lead to therapies for the treatment of PE.
Collapse
Affiliation(s)
- Kelsey R Bounds
- Department of Medical Physiology, Texas A&M Health Science Center , Temple, TX , USA
| | | | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M Health Science Center , Temple, TX , USA
| |
Collapse
|
28
|
Ueki N, Takeda S, Koya D, Kanasaki K. The relevance of the Renin-Angiotensin system in the development of drugs to combat preeclampsia. Int J Endocrinol 2015; 2015:572713. [PMID: 26000015 PMCID: PMC4426891 DOI: 10.1155/2015/572713] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 03/28/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia is a hypertensive disorder that occurs during pregnancy. It has an unknown etiology and affects approximately 5-8% of pregnancies worldwide. The pathophysiology of preeclampsia is not yet known, and preeclampsia has been called "a disease of theories." The central symptom of preeclampsia is hypertension. However, the etiology of the hypertension is unknown. In this review, we analyze the molecular mechanisms of preeclampsia with a particular focus on the pathogenesis of the hypertension in preeclampsia and its association with the renin-angiotensin system. In addition, we propose potential alternative strategies to target the renin-angiotensin system, which is enhanced during pregnancy.
Collapse
Affiliation(s)
- Norikazu Ueki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa 920-0293, Japan
- Department of Obstetrics and Gynecology, Juntendo University, Tokyo 113-8431, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University, Tokyo 113-8431, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa 920-0293, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Ishikawa 920-0293, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
- *Keizo Kanasaki:
| |
Collapse
|
29
|
Kobayashi Y, Yamamoto T, Chishima F, Takahashi H, Suzuki M. Autoantibodies isolated from patients with preeclampsia induce soluble endoglin production from trophoblast cells via interactions with angiotensin II type 1 receptor. Am J Reprod Immunol 2014; 73:285-91. [PMID: 25376533 DOI: 10.1111/aji.12340] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/14/2014] [Indexed: 11/29/2022] Open
Abstract
PROBLEM This study investigated whether angiotensin II type 1 receptor agonistic autoantibodies (AT1 -AAs) mediate the increased release of soluble endoglin (sEng) in women with preeclampsia. METHOD OF STUDY Serum samples were obtained from women with normal pregnancies or with preeclampsia. Human first-trimester trophoblast cells were cultured with purified IgG derived from these sera, and the sEng protein and mRNA expression levels were measured in the supernatants. We also determined the effects of the AT1 -AAs on these cells following treatment with an AT1 receptor antagonist (losartan). RESULTS Compared with the IgG isolated from the women with normal pregnancies, treatments of the preeclamptic patients markedly increased sEng production and mRNA expression in trophoblast cells. Co-treatment with losartan significantly attenuated the release of sEng and sEng mRNA expression in the trophoblast cells. CONCLUSION AT1 -AAs may be related to the increased release of sEng observed during preeclampsia and may play important roles in the pathology of this disorder.
Collapse
Affiliation(s)
- Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
30
|
Minuz P, Fava C, Hao S, Pedraza P, Amen G, Meneguzzi A, Vattemi G, Marini M, Zanconato G, Ferreri NR. Differential regulation of TNF receptors in maternal leukocytes is associated with severe preterm preeclampsia. J Matern Fetal Neonatal Med 2014; 28:869-75. [PMID: 25034210 DOI: 10.3109/14767058.2014.937695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We tested the hypothesis that maternal peripheral blood leukocytes contribute to elevated levels of soluble TNF receptors (sTNFR) in preeclampsia (PE) with concomitant intrauterine growth restriction (IUGR). TNFR1 and TNFR2 were evaluated in a cross-sectional study comparing preeclamptic (n = 15) with or without IUGR versus normotensive pregnant women (PREG, n = 30), and non-pregnant controls (Con; n = 20). Plasma levels of sTNFR1 were higher in PE (1675.0 ± 227.1 pg/mL) compared with PREG (1035.0 ± 101.1 pg/mL) and Con (589.3 ± 82.67 pg/mL), with the highest values observed in PE with IUGR (2624.0 ± 421.4 pg/mL; n = 6). Plasma sTNFR2 was higher during pregnancy (PE: 1836.0 ± 198.7 pg/mL; PREG: 1697.0 ± 95.0 pg/mL) compared with Con (598.3 ± 82.7 pg/mL). Urinary levels of sTNFR1 and sTNFR2 were higher in PE and PREG compared with the Con group. Abundance of TNFR1 mRNA in peripheral blood leukocytes was strongly correlated with plasma levels of sTNFR1 in PE. However, TNFR2 mRNA accumulation in leukocytes did not correlate with sTNFR2 plasma levels. The level of sTNFR1 in plasma was correlated with body weight of the newborn (r = -0.56). The data suggest that maternal leukocytes contribute to sTNFR1 levels in plasma in association with decreasing newborn weight and PE with concomitant IUGR.
Collapse
Affiliation(s)
- Pietro Minuz
- Department of Pharmacology, New York Medical College , Valhalla, NY , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Exposure to AT1 receptor autoantibodies during pregnancy increases susceptibility of the maternal heart to postpartum ischemia-reperfusion injury in rats. Int J Mol Sci 2014; 15:11495-509. [PMID: 24979132 PMCID: PMC4139795 DOI: 10.3390/ijms150711495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/04/2014] [Accepted: 06/10/2014] [Indexed: 01/26/2023] Open
Abstract
Epidemiological studies have demonstrated that women with a history of preeclampsia have a two-fold increased risk of developing cardiovascular diseases in later life. It is not known whether or not this risk is associated with angiotensin II receptor type 1 autoantibody (AT1-AA), an agonist acting via activation of AT1 receptor (AT1R), which is believed to be involved in the pathogenesis of preeclampsia. The objective of the present study was to confirm the hypothesis that AT1-AA exposure during pregnancy may change the maternal cardiac structure and increase the susceptibility of the postpartum heart to ischemia/reperfusion injury (IRI). In the present study, we first established a preeclampsia rat model by intravenous injection of AT1-AA extracted from the plasma of rats immunized with AT1R, observed the susceptibility of the postpartum maternal heart to IRI at 16 weeks postpartum using the Langendorff preparation, and examined the cardiac structure using light and transmission electron microscopy. The modeled animals presented with symptoms very similar to the clinical symptoms of human preeclampsia during pregnancy, including hypertension and proteinuria. The left ventricular weight (LVW) and left ventricular mass index (LVMI) in AT1-AA treatment group were significantly increased as compared with those of the control group (p < 0.01), although there was no significant difference in final weight between the two groups. AT1-AA acting on AT1R not only induced myocardial cell hypertrophy, mitochondrial swelling, cristae disorganization and collagen accumulation in the interstitium but affected the left ventricular (LV) function and delayed recovery from IRI. In contrast, co-treatment with AT1-AA + losartan completely blocked AT1-AA-induced changes in cardiac structure and function. These data indicate that the presence of AT1-AA during pregnancy was strongly associated with the markers of LV geometry changes and remodeling, and increased the cardiac susceptibility to IRI in later life of postpartum maternal rats.
Collapse
|
32
|
Ramma W, Ahmed A. Therapeutic potential of statins and the induction of heme oxygenase-1 in preeclampsia. J Reprod Immunol 2014; 101-102:153-160. [PMID: 24503248 PMCID: PMC4003533 DOI: 10.1016/j.jri.2013.12.120] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 11/28/2013] [Accepted: 12/20/2013] [Indexed: 02/05/2023]
Abstract
Heme oxygenase (Hmox) is an endogenous system that offers protection against placental cytotoxic damage associated with preeclampsia. The Hmox1/carbon monoxide (CO) pathway inhibits soluble Flt-1 (sFlt-1) and soluble Endoglin (sEng). More importantly, statins induce Hmox1 and suppress the release of sFlt-1 and sEng; thus, statins and Hmox1 activators are potential novel therapeutic agents for treating preeclampsia. The contribution of the Hmox system to the pathogenesis of preeclampsia has been further indicated by the incidence of preeclampsia being reduced by a third in smokers, who had reduced levels of circulating sFlt-1. Interestingly, preeclamptic women exhale less CO compared with women with healthy pregnancies. Hmox1 is reduced prior to the increase in sFlt-1 as Hmox1 mRNA expression in the trophoblast is decreased in the first trimester in women who go on to develop preeclampsia. Induction of Hmox1 or exposure to CO or bilirubin has been shown to inhibit the release of sFlt-1 and sEng in animal models of preeclampsia. The functional benefit of statins and Hmox1 induction in women with preeclampsia is valid not only because they inhibit sFlt-1 release, but also because statins and Hmox1 are associated with anti-apoptotic, anti-inflammatory, and anti-oxidant properties. The StAmP trial is the first randomized control trial (RCT) evaluating the use of pravastatin to ameliorate severe preeclampsia. This proof-of-concept study will pave the way for future global RCT, the success of which will greatly contribute to achieving the United Nations Millennium Development Goals (MDG4 and MDG5) and offering an affordable and easily accessible therapy for preeclampsia.
Collapse
Affiliation(s)
- Wenda Ramma
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Asif Ahmed
- Vascular Biology Laboratory, School of Medical Sciences, Aston University, Birmingham B4 7ET, England, United Kingdom
| |
Collapse
|
33
|
George EM, Granger JP. Recent insights into the pathophysiology of preeclampsia. ACTA ACUST UNITED AC 2014; 5:557-566. [PMID: 21170149 DOI: 10.1586/eog.10.45] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Preeclampsia, characterized by new-onset gestational hypertension and proteinuria, is a common and serious complication of pregnancy. Evidence from both animal and human studies has implicated placental ischemia and hypoxia as a central causative factor in the etiology of the disorder. The ischemic placenta in turn initiates a cascade of secondary effector mechanisms, including altered proangiogenic and antiangiogenic factor balance, increase in maternal oxidative stress and endothelial and immunological dysfunction. The full elucidation of these mechanisms will hopefully lead to a more complete understanding of the etiology of preeclampsia and lead to successful therapeutic intervention through the targeted disruption of new and novel pathways.
Collapse
Affiliation(s)
- Eric M George
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | | |
Collapse
|
34
|
Abstract
Stimulating antibodies against G-protein-coupled receptors, including the β1- and β2-adrenergic receptors, the α1-adrenergic receptor, and the angiotensin II AT1 receptor, have been described, as well as activating antibodies directed at the platelet-derived growth factor receptor tyrosine kinase. Their existence and actions appear to be established. Lacking are mechanistic studies of receptor activation and translational studies to document receptor-stimulating antibodies as worthwhile therapeutic targets.
Collapse
Affiliation(s)
- Friedrich C Luft
- Experimental and Clinical Research Center and Max-Delbrück Center for Molecular Medicine and Charité Medical Faculty, Berlin, Germany.
| |
Collapse
|
35
|
Genest DS, Falcao S, Michel C, Kajla S, Germano MF, Lacasse AA, Vaillancourt C, Gutkowska J, Lavoie JL. Novel role of the renin-angiotensin system in preeclampsia superimposed on chronic hypertension and the effects of exercise in a mouse model. Hypertension 2013; 62:1055-61. [PMID: 24101664 DOI: 10.1161/hypertensionaha.113.01983] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gestational hypertensive disorders, such as preeclampsia, affect 6% to 8% of all pregnancies in North America, and they are the leading cause of maternal mortality in industrialized countries, accounting for 16% of deaths. Women with hypertension have an increased risk (15% to 25%) of developing preeclampsia. Our aim was to investigate the mechanisms implicated in preeclampsia superimposed on chronic hypertension and in the protective effects of exercise in a mouse model. Female mice overexpressing human angiotensinogen and human renin were used as a model of preeclampsia superimposed on chronic hypertension. In the trained group, mothers were placed in cages with access to a wheel before mating, and they remained within these throughout gestation. Blood pressure was measured by telemetry. We found that angiotensin II type I receptor was increased, whereas the Mas receptor was decreased in the placenta and the aorta of pregnant sedentary transgenic mice. This would produce a decrease in angiotensin-(1-7) effects in favor of angiotensin II. Supporting the functional contribution of this modulation, we found that the prevention of most pathological features in trained transgenic mice was associated with a normalization of placental angiotensin II type 1 and Mas receptors and an increase in aortic Mas receptor. We also found reduced circulating and placental soluble Fms-like tyrosine kinase-1 in trained transgenic mice compared with sedentary mice. This study demonstrates that modulation of the renin-angiotensin system is a key mechanism in the development of preeclampsia superimposed on chronic hypertension, which can be altered by exercise training to prevent disease features in an animal model.
Collapse
Affiliation(s)
- Dominique S Genest
- CRCHUM - Technopôle Angus, 2901 Rachel St E, Suite 310, Montreal, Quebec H1W 4A4, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fenton C, Hobson SR, Wallace EM, Lim R. Future therapies for pre-eclampsia: beyond treading water. Aust N Z J Obstet Gynaecol 2013; 54:3-8. [DOI: 10.1111/ajo.12134] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 08/09/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Christine Fenton
- The Ritchie Centre; Monash Institute of Medical Research; Melbourne Victoria Australia
| | - Sebastian R. Hobson
- Department of Obstetrics and Gynaecology; Monash University; Melbourne Victoria Australia
| | - Euan M. Wallace
- The Ritchie Centre; Monash Institute of Medical Research; Melbourne Victoria Australia
- Department of Obstetrics and Gynaecology; Monash University; Melbourne Victoria Australia
| | - Rebecca Lim
- The Ritchie Centre; Monash Institute of Medical Research; Melbourne Victoria Australia
- Department of Obstetrics and Gynaecology; Monash University; Melbourne Victoria Australia
| |
Collapse
|
37
|
Abstract
Hypertensive disorders are life-threatening diseases with high morbidity and mortality, affecting billions of individuals worldwide. A multitude of underlying conditions may contribute to hypertension, thus the need for a plethora of treatment options to identify the approach that best meets the needs of individual patients. A growing body of evidence indicates that (1) autoantibodies that bind to and activate the major angiotensin II type I (AT₁) receptor exist in the circulation of patients with hypertensive disorders, (2) these autoantibodies contribute to disease pathophysiology, (3) antibody titers correlate to the severity of the disease, and (4) efforts to block or remove these pathogenic autoantibodies have therapeutic potential. These autoantibodies, termed AT₁ agonistic autoantibodies have been extensively characterized in preeclampsia, a life-threatening hypertensive condition of pregnancy. As reviewed here, these autoantibodies cause symptoms of preeclampsia when injected into pregnant mice. Somewhat surprisingly, these auto antibodies also appear in 3 animal models of preeclampsia. However, the occurrence of AT₁ agonistic autoantibodies is not restricted to pregnancy. These autoantibodies are prevalent among kidney transplant recipients who develop severe transplant rejection and malignant hypertension during the first week after transplantation. AT₁ agonistic autoantibodies are also highly abundant among a group of patients with essential hypertension that are refractory to standard therapy. More recently these autoantibodies have been seen in patients with the autoimmune disease, systemic sclerosis. These 3 examples extend the clinical impact of AT₁ agonistic autoantibodies beyond pregnancy. Research reviewed here raises the intriguing possibility that preeclampsia and other hypertensive conditions are autoimmune diseases characterized by the presence of pathogenic autoantibodies that activate the major angiotensin receptor, AT₁. These pathogenic autoantibodies could serve as presymptomatic biomarkers and therapeutic targets, thereby providing improved medical management for these conditions.
Collapse
Affiliation(s)
- Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | | |
Collapse
|
38
|
Liu NQ, Ouyang Y, Bulut Y, Lagishetty V, Chan SY, Hollis BW, Wagner C, Equils O, Hewison M. Dietary vitamin D restriction in pregnant female mice is associated with maternal hypertension and altered placental and fetal development. Endocrinology 2013; 154:2270-80. [PMID: 23677931 DOI: 10.1210/en.2012-2270] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epidemiology has linked vitamin D deficiency with preeclampsia in humans. We hypothesized that low vitamin D status in pregnant mice may lead to symptoms of preeclampsia. Female BL6 mice were raised on vitamin D-sufficient or -deficient diets from weeks 4 of age and then mated with vitamin D-sufficient BL6 males at week 8. The resulting pregnant mice were either allowed to deliver pups and monitored for blood pressure (BP) and weight of offspring or euthanized at day 14 or 18 of gestation (E14 or E18) for analysis of serum, placental/kidney tissues, and fetuses. At E14 serum concentrations of 25-hydroxyvitamin D (30.1 ± 5.0 vs 1.8 ± 0.6 ng/mL, P < .001) and 1,25-dihydroxyvitamin D (119.5 ± 18.7 vs 37.4 ± 5.1 pg/mL, P < .01) were higher in sufficient vs deficient pregnant mice. At E14 BP was significantly elevated in vitamin D-deficient pregnant mice relative to vitamin D-sufficient mice for both systolic BP (124.89 ± 2.28 vs 105.34 ± 3.61 mm Hg, P < .001) and mean arterial pressure (115.33 ± 1.93 vs 89.33 ± 5.02 mm Hg, P < .001). This elevation continued through pregnancy until 7 days postpartum (PP7) but returned to baseline by PP14. Analysis of maternal kidneys showed increased expression of mRNA for renin and the angiotensin II receptor (3- and 4-fold, respectively) in vitamin D-deficient vs -sufficient mice at E14. Histological analysis of E14 placentas from vitamin D-deficient mice showed decreased vascular diameter within the labyrinth region. E14 and E18 fetuses from vitamin D-deficient mice were larger than those from vitamin D-sufficient mothers. However, by PP14 pups from vitamin D-deficient mothers weighed significantly less than those from vitamin D-sufficient mothers. Resupplementation of vitamin D periconceptually partially reversed the effects of vitamin D deficiency. These data provide further evidence that low vitamin D status may predispose pregnant women to dysregulated placental development and elevated blood pressure.
Collapse
Affiliation(s)
- Nancy Q Liu
- Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zenclussen AC. Adaptive immune responses during pregnancy. Am J Reprod Immunol 2013; 69:291-303. [PMID: 23418773 DOI: 10.1111/aji.12097] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 01/23/2013] [Indexed: 02/05/2023] Open
Abstract
It has long been believed that there is no immune interaction between mother and conceptus during pregnancy. This concept changed after evidence was provided that the maternal immune system is aware of the semiallogeneic conceptus and develops strategies to tolerate it. Since then, finely regulated mechanisms of active tolerance toward the fetus have been described. This Special Issue of the American Journal of Reproductive Immunology deals with these mechanisms. It begins with the description of minor histocompatibility antigens in the placenta; it further goes through adaptive immune responses toward paternal fetal antigens, mostly concentrating on regulatory T cells and molecules modulating the Th1/Th2 balance. The participation of antibody-producing B cells in normal and pathological pregnancies is also discussed. This introductory chapter resumes the concepts presented throughout the Issue and discusses the clinical applications raised from these concepts.
Collapse
Affiliation(s)
- Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
40
|
Chen X, Qiu Z, Yang S, Ding D, Chen F, Zhou Y, Wang M, Lin J, Yu X, Zhou Z, Liao Y. Effectiveness and Safety of a Therapeutic Vaccine Against Angiotensin II Receptor Type 1 in Hypertensive Animals. Hypertension 2013. [DOI: 10.1161/hypertensionaha.112.201020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiao Chen
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Qiu
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Yang
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dan Ding
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Fen Chen
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yanzhao Zhou
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jibin Lin
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xian Yu
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yuhua Liao
- From the Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of the Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
RAS in Pregnancy and Preeclampsia and Eclampsia. Int J Hypertens 2012; 2012:739274. [PMID: 23346385 PMCID: PMC3546487 DOI: 10.1155/2012/739274] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 12/14/2012] [Indexed: 11/18/2022] Open
Abstract
Preeclampsia is a common disease of pregnancy characterized by the presence of hypertension and commitment of many organs, including the brain, secondary to generalized endothelial dysfunction. Its etiology is not known precisely, but it involved several factors, highlighting the renin angiotensin system (RAS), which would have an important role in the origin of multisystem involvement. This paper reviews the evidence supporting the involvement of RAS in triggering the disease, in addition to the components of this system that would be involved and how it eventually produces brain engagement.
Collapse
|
42
|
Herse F, LaMarca B. Angiotensin II type 1 receptor autoantibody (AT1-AA)-mediated pregnancy hypertension. Am J Reprod Immunol 2012; 69:413-8. [PMID: 23279165 DOI: 10.1111/aji.12072] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/06/2012] [Indexed: 11/28/2022] Open
Abstract
Autoantibodies can cause complications in pregnancy. Preeclampsia is the leading cause of maternal and fetal morbidity and mortality during pregnancy. Overall, 5-10% of all pregnancies worldwide develop preeclampsia. Women who developed preeclampsia and their children have an increased risk to suffer from cardiovascular diseases later in life. In preeclampsia, agonistic autoantibodies against the angiotensin II type 1 receptor autoantibodies (AT1-AA) are described. They induce NADPH oxidase and the MAPK/ERK pathway leading to NF-κB and tissue factor activation. AT1-AA are detectable in animal models of preeclampsia and are responsible for elevation of soluble fms-related tyrosine kinase-1 (sFlt1) and soluble endoglin (sEng), oxidative stress, and endothelin-1, all of which are enhanced in preeclamptic women. AT1-AA can be detected in pregnancies with abnormal uterine perfusion and increased resistance index as well as in patients with systemic sclerosis and renal allograft rejection. This review discusses the current knowledge about the AT1-AA, its signaling, and their impact in pregnancy complications and other autoimmune disorders.
Collapse
Affiliation(s)
- Florian Herse
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
43
|
Wang W, Irani RA, Zhang Y, Ramin SM, Blackwell SC, Tao L, Kellems RE, Xia Y. Autoantibody-mediated complement C3a receptor activation contributes to the pathogenesis of preeclampsia. Hypertension 2012; 60:712-21. [PMID: 22868393 DOI: 10.1161/hypertensionaha.112.191817] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preeclampsia (PE) is a prevalent life-threatening hypertensive disorder of pregnancy associated with increased complement activation. However, the causative factors and pathogenic role of increased complement activation in PE are largely unidentified. Here we report that a circulating maternal autoantibody, the angiotensin II type 1 receptor agonistic autoantibody, which emerged recently as a potential pathogenic contributor to PE, stimulates deposition of complement C3 in placentas and kidneys of pregnant mice via angiotensin II type 1 receptor activation. Next, we provide in vivo evidence that selectively interfering with C3a signaling by a complement C3a receptor-specific antagonist significantly reduces hypertension from 167±7 to 143±5 mm Hg and proteinuria from 223.5±7.5 to 78.8±14.0 μg of albumin per milligram creatinine (both P<0.05) in angiotensin II type 1 receptor agonistic autoantibody-injected pregnant mice. In addition, we demonstrated that complement C3a receptor antagonist significantly inhibited autoantibody-induced circulating soluble fms-like tyrosine kinase 1, a known antiangiogenic protein associated with PE, and reduced small placental size with impaired angiogenesis and intrauterine growth restriction. Similarly, in humans, we demonstrate that C3 deposition is significantly elevated in the placentas of preeclamptic patients compared with normotensive controls. Lastly, we show that complement C3a receptor activation is a key mechanism underlying autoantibody-induced soluble fms-like tyrosine kinase 1 secretion and decreased angiogenesis in cultured human villous explants. Overall, we provide mouse and human evidence that angiotensin II type 1 receptor agonistic autoantibody-mediated activation contributes to elevated C3 and that complement C3a receptor signaling is a key mechanism underlying the pathogenesis of the disease. These studies are the first to link angiotensin II type 1 receptor agonistic autoantibody with complement activation and to provide important new opportunities for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Wei Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Hunan, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Soluble fms-like tyrosine kinase 1 and soluble endoglin are elevated circulating anti-angiogenic factors in pre-eclampsia. Pregnancy Hypertens 2012; 2:358-67. [PMID: 26105603 DOI: 10.1016/j.preghy.2012.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/24/2012] [Indexed: 01/05/2023]
Abstract
Pre-eclampsia, characterized by hypertension and proteinuria, affects approximately 3-5% of all pregnancies worldwide and is a major cause of maternal and fetal morbidity and mortality. Maternal endothelial dysfunction is associated with disease pathogenesis. Recently, reports have shown that elevated levels of circulating soluble fms-like tyrosine kinase 1 [sFlt1] and soluble endoglin [sEng] are associated with pre-eclampsia. Flt1 is a receptor for vascular endothelial growth factor receptor [VEGF], whereas endoglin [Eng] is an auxiliary receptor for transforming growth factor-β [TGF-β] super-family members. Both signaling pathways modulate angiogenesis and are involved in vascular homeostasis. Increased levels of sFlt1 and sEng dysregulate VEGF and TGF-β signaling respectively, resulting in endothelial dysfunction of maternal blood vessels. This review summarizes our current knowledge of Flt1 and endoglin and soluble forms in pre-eclampsia. Furthermore, it highlights the predictive and early-screening value of circulating levels of sFlt1 and sEng for the risk of developing pre-eclampsia.
Collapse
|
45
|
Jensen F, Wallukat G, Herse F, Budner O, El-Mousleh T, Costa SD, Dechend R, Zenclussen AC. CD19
+
CD5
+
Cells as Indicators of Preeclampsia. Hypertension 2012; 59:861-8. [DOI: 10.1161/hypertensionaha.111.188276] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preeclampsia is a devastating pregnancy-associated disorder affecting 5% to 8% of pregnant women worldwide. It emerges as an autoimmune-driven disease, and, among others, the autoantibodies against angiotensin type 1 receptor II have been proposed to account for preeclampsia symptoms. Despite much attention focused on describing autoantibodies associated with preeclampsia, there is no clue concerning the cell population producing them. CD19
+
CD5
+
B-1a B cells constitute the main source of natural and polyreactive antibodies, which can be directed against own structures. Here, we aimed to identify the B-cell subpopulation responsible for autoantibody production during preeclampsia and to study their regulation, as well as their possible use as markers for the disease. The frequency of CD19
+
CD5
+
cells in peripheral blood of preeclamptic patients is dramatically increased compared with normal pregnant women as analyzed by flow cytometry. This seems to be driven by the high human chorionic gonadotropin levels present in the serum and placenta supernatant of preeclamptic patients versus normal pregnant women. Not only ≈95% of CD19
+
CD5
+
cells express the human chorionic gonadotropin receptor, but these cells also expand on human chorionic gonadotropin stimulation in a lymphocyte culture. Most importantly, isolated CD19
+
CD5
+
cells produce autoantibodies against angiotensin type 1 receptor II, and CD19
+
CD5
+
cells were further detected in the placenta of preeclamptic but not of normal pregnancies where barely B cells are present. Our results identify a B-cell population able to produce pregnancy-pathological autoantibodies as possible markers for preeclampsia, which opens vast diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Federico Jensen
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Gerd Wallukat
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Florian Herse
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Oliver Budner
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Tarek El-Mousleh
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Serban-Dan Costa
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Ralf Dechend
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| | - Ana Claudia Zenclussen
- From the Experimental Obstetrics and Gynecology (F.J., T.E.-M., A.C.Z.), Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Experimental and Clinical Research Center (G.W., F.H., R.D.), a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany; University Women's Clinic (O.B., S.-D.C.), Otto-von-Guericke University, Magdeburg, Germany; HELIOS Klinikum Berlin-Buch (R.D.), Berlin, Germany
| |
Collapse
|
46
|
Xia Y, Kellems RE. Receptor-activating autoantibodies and disease: preeclampsia and beyond. Expert Rev Clin Immunol 2011; 7:659-74. [PMID: 21895478 DOI: 10.1586/eci.11.56] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The research reviewed in this article provides examples of autoantibody-mediated receptor activation that likely contributes to disease. The classic example is Graves' hyperthyroidism, in which autoantibodies activate the thyroid-stimulating hormone receptor resulting in overproduction of thyroid hormones. Other compelling examples come from the cardiovascular literature and include agonistic autoantibodies targeting the cardiac β(1)-adrenergic receptor, which are associated with dilated cardiomyopathy. Autoantibodies capable of activating α(1)-adrenergic receptors are associated with refractory hypertension and cardiomyopathy. A prominent example is preeclampsia, a hypertensive disease of pregnancy, characterized by the presence of autoantibodies that activate the major angiotensin receptor, AT(1). AT(1) receptor-activating autoantibodies are also observed in kidney transplant recipients suffering from severe vascular rejection and malignant hypertension. AT(1) receptor-activating autoantibodies and antibodies that activate the endothelin-1 receptor, ET(A), are prevalent in individuals diagnosed with systemic sclerosis. Thus, the presence of agonistic autoantibodies directed to G protein-coupled receptors has been observed in numerous cardiovascular disease states. Rapidly emerging evidence indicates that receptor-activating autoantibodies contribute to disease, and that efforts to detect and remove these pathogenic autoantibodies or block their actions will provide promising therapeutic possibilities.
Collapse
Affiliation(s)
- Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School, Houston, TX 77030, USA.
| | | |
Collapse
|
47
|
Zhang W, Wang W, Yu H, Zhang Y, Dai Y, Ning C, Tao L, Sun H, Kellems RE, Blackburn MR, Xia Y. Interleukin 6 underlies angiotensin II-induced hypertension and chronic renal damage. Hypertension 2011; 59:136-44. [PMID: 22068875 DOI: 10.1161/hypertensionaha.111.173328] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic kidney disease (CKD) is a prevalent life-threatening disease frequently associated with hypertension, progression to renal fibrosis, and eventual renal failure. Although the pathogenesis of CKD remains largely unknown, an increased inflammatory response is known to be associated with the disease and has long been speculated to contribute to disease development. However, the causative factors, the exact role of the increased inflammatory cascade in CKD, and the underlying mechanisms for its progression remain unidentified. Here we report that interleukin 6 (IL-6) expression levels were significantly increased in the kidneys collected from CKD patients and further elevated in CKD patients characterized with hypertension. Functionally, we determined that angiotensin II is a causative factor responsible for IL-6 induction in the mouse kidney and that genetic deletion of IL-6 significantly reduced hypertension and key features of CKD, including renal injury and progression to renal fibrosis in angiotensin II-infused mice. Mechanistically, we provide both human and mouse evidence that IL-6 is a key cytokine functioning downstream of angiotensin II signaling to directly induce fibrotic gene expression and preproendothelin 1 mRNA expression in the kidney. Overall, both the mouse and human studies reported here provide evidence that angiotensin II induces IL-6 production in the kidney, and that, in addition to its role in hypertension, increased IL-6 may play an important pathogenic role in CKD by inducing fibrotic gene expression and ET-1 gene expression. These findings immediately suggest that the IL-6 signaling is a novel therapeutic target to manage this devastating disorder affecting millions worldwide.
Collapse
Affiliation(s)
- Weiru Zhang
- Department of Biochemistry, University of Texas-Houston Medical School, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Maganto-García E, Bu DX, Tarrio ML, Alcaide P, Newton G, Griffin GK, Croce KJ, Luscinskas FW, Lichtman AH, Grabie N. Foxp3+-inducible regulatory T cells suppress endothelial activation and leukocyte recruitment. THE JOURNAL OF IMMUNOLOGY 2011; 187:3521-9. [PMID: 21873519 DOI: 10.4049/jimmunol.1003947] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The ability of regulatory T cells (Treg) to traffic to sites of inflammation supports their role in controlling immune responses. This feature supports the idea that adoptive transfer of in vitro expanded human Treg could be used for treatment of immune/inflammatory diseases. However, the migratory behavior of Treg, as well as their direct influence at the site of inflammation, remains poorly understood. To explore the possibility that Treg may have direct anti-inflammatory influences on tissues, independent of their well-established suppressive effects on lymphocytes, we studied the adhesive interactions between mouse Treg and endothelial cells, as well as their influence on endothelial function during acute inflammation. We show that Foxp3(+) adaptive/inducible Treg (iTreg), but not naturally occurring Treg, efficiently interact with endothelial selectins and transmigrate through endothelial monolayers in vitro. In response to activation by endothelial Ag presentation or immobilized anti-CD3ε, Foxp3(+) iTreg suppressed TNF-α- and IL-1β-mediated endothelial selectin expression and adhesiveness to effector T cells. This suppression was contact independent, rapid acting, and mediated by TGF-β-induced activin receptor-like kinase 5 signaling in endothelial cells. In addition, Foxp3(+) iTreg adhered to inflamed endothelium in vivo, and their secretion products blocked acute inflammation in a model of peritonitis. These data support the concept that Foxp3(+) iTreg help to regulate inflammation independently of their influence on effector T cells by direct suppression of endothelial activation and leukocyte recruitment.
Collapse
|
49
|
Abstract
Preeclampsia, a hypertensive disorder peculiar to pregnancy, is a systemic syndrome that appears to originate in the placenta and is characterized by widespread maternal endothelial dysfunction. Until recently, the molecular pathogenesis of phenotypic preeclampsia was largely unknown, but recent observations support the hypothesis that altered expression of placental anti-angiogenic factors are responsible for the clinical manifestations of the disease. Soluble Flt1 and soluble endoglin, secreted by the placenta, are increased in the maternal circulation weeks before the onset of preeclampsia. These anti-angiogenic factors produce systemic endothelial dysfunction, resulting in hypertension, proteinuria, and the other systemic manifestations of preeclampsia. The molecular basis for placental dysregulation of these pathogenic factors remains unknown, and as of 2011 the role of angiogenic proteins in early placental vascular development was starting to be explored. The data linking angiogenic factors to preeclampsia have exciting clinical implications, and likely will transform the detection and treatment of preeclampsia.
Collapse
Affiliation(s)
- Sharon E Maynard
- Department of Medicine, Division of Renal Diseases and Hypertension, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| | | |
Collapse
|
50
|
Abstract
Many reports indicate that there is an increase in almost all of the components of the renin-angiotensin system (RAS) during an uncomplicated pregnancy, but renin activity, angiotensin II, and aldosterone decrease in preeclampsia (PE) for reasons that are unclear. PE is a life-threatening disorder of late pregnancy characterized by hypertension, proteinuria, increased soluble fms-like tyrosine kinase-1, as well as renal and placental morphologic abnormalities. Although a leading cause of maternal and perinatal morbidity and mortality, the pathogenic mechanisms of PE remain largely undefined. Immunologic mechanisms and aberrations of the RAS have been long considered contributors to the disorder. Bridging these two concepts, numerous studies report the presence of the angiotensin II type I receptor agonistic autoantibody (AT(1)-AA) found circulating in preeclamptic women. This autoantibody induces many key features of the disorder through AT(1) receptor signaling, and has been implicated in the pathogenesis of PE. Here we review the functions of the RAS during normal pregnancy and PE, and highlight the role of AT(1)-AA in both animal models and in the human disorder.
Collapse
Affiliation(s)
- Roxanna A Irani
- Department of Biochemistry & Molecular Biology, University of Texas at Houston Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | | |
Collapse
|