151
|
Taylor EB, Wolf VL, Dent E, Ryan MJ. Mechanisms of hypertension in autoimmune rheumatic diseases. Br J Pharmacol 2019; 176:1897-1913. [PMID: 30714094 PMCID: PMC6534791 DOI: 10.1111/bph.14604] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023] Open
Abstract
Patients with autoimmune rheumatic diseases including rheumatoid arthritis and systemic lupus erythematosus have an increased prevalence of hypertension. There is now a large body of evidence showing that the immune system is a key mediator in both human primary hypertension and experimental models. Many of the proposed immunological mechanisms leading to primary hypertension are paralleled in autoimmune rheumatic disorders. Therefore, examining the link between autoimmunity and hypertension can be informative for understanding primary hypertension. This review examines the prevalent hypertension, the immune mediators that contribute to the prevalent hypertension and their impact on renal function and how the risk of hypertension is potentially influenced by common hormonal changes that are associated with autoimmune rheumatic diseases. Linked Articles This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Victoria L Wolf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Elena Dent
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA.,G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
152
|
Wang Z, Wu Y, Zhang S, Zhao Y, Yin X, Wang W, Ma X, Liu H. The role of NO-cGMP pathway inhibition in vascular endothelial-dependent smooth muscle relaxation disorder of AT1-AA positive rats: protective effects of adiponectin. Nitric Oxide 2019; 87:10-22. [PMID: 30831264 DOI: 10.1016/j.niox.2019.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/16/2019] [Accepted: 02/19/2019] [Indexed: 12/24/2022]
Abstract
Angiotensin II type 1 receptor autoantibodies (AT1-AA) cause endothelial-dependent smooth muscle relaxation disorder. It is well understood that impairment of the NO-cGMP signaling pathway is one of the mechanisms of endothelial-dependent smooth muscle relaxation disorder. However, it is still unclear whether AT1-AA induces endothelial-dependent smooth muscle relaxation disorder via the impairment of the NO-cGMP signaling pathway. In addition, adiponectin exerts vascular endothelial protection through the NO-cGMP signaling pathway. Therefore, the purpose of this investigation was to assess the mechanism of vascular endothelial-dependent smooth muscle relaxation disorder induced by AT1-AA and the role of adiponectin in attenuating this dysregulation. Serum endothelin-1 (ET-1), adiponectin and AT1-AA were detected by enzyme-linked immunosorbent assay. In preeclamptic patients, there was an increased level of AT1-AA, which was positively correlated with ET-1 and negatively correlated with adiponectin, as elevated levels of ET-1 suggested endothelial injury. AT1-AA-positive animal models were actively immunized with the second extracellular loop of the angiotensin II type 1 receptor (AT1R-ECII) for eight weeks. In thoracic aortas of AT1-AA positive rats, ET-1 was elevated, endothelium-dependent vasodilation was decreased. Paradoxically, as the upstream element of the NO-cGMP signaling pathway, NO production was not decreased but increased, and the ratio of p-VASP/VASP, an established biochemical endpoint of NO-cGMP signaling pathway, was reduced. Moreover, the levels of nitrotyrosine and gp91phox which indicate the presence of peroxynitrite (ONOO-) and superoxide anion (O2·-) were increased. Pretreatment with the ONOO- scavenger FeTMPyP or O2·-scavenger Tempol normalized vasorelaxation. Key enzymes responsible for NO synthesis were also assessed. iNOS protein expression was increased, but p-eNOS(Ser1177)/eNOS was reduced. Preincubation with the iNOS inhibitor 1400 W or eNOS agonist nebivolol restored vasorelaxation. Further experiments showed levels of p-AMPKα (Thr172)/AMPKα, which controls iNOS expression and eNOS activity, to have been reduced. Furthermore, levels of the upstream component of AMPK, adiponectin, was reduced in sera of AT1-AA positive rats and supplementation of adiponectin significantly decreased ET-1 contents, improved endothelial-dependent vasodilation, reduced NO production, elevated p-VASP/VASP, inhibited protein expression of nitrotyrosine and gp91phox, reduced iNOS overexpression, and increased eNOS phosphorylation at Ser1177 in the thoracic aorta of AT1-AA positive rats. These results established that impairment NO-cGMP pathway may aggravate the endothelial-dependent smooth muscle relaxation disorder in AT1-AA positive rats and adiponectin improved endothelial-dependent smooth muscle relaxation disorder by enhancing NO-cGMP pathway. This discovery may shed a novel light on clinical treatment of vascular diseases associated with AT1-AA.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Yuhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Xiaochen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Xinliang Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China; Department of Emergency Medicine, Thomas Jefferson University, Philadephia, Pennsylvania, USA.
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
153
|
Campbell N, LaMarca B, Cunningham MW. The Role of Agonistic Autoantibodies to the Angiotensin II Type 1 Receptor (AT1-AA) in Pathophysiology of Preeclampsia. Curr Pharm Biotechnol 2019; 19:781-785. [PMID: 30255752 DOI: 10.2174/1389201019666180925121254] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 07/20/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022]
Abstract
Preeclampsia is the leading cause of death and morbidity worldwide for the mother and fetus during pregnancy. Preeclampsia does not only affect the mother and the baby during pregnancy, but can also have long-term effects, such as the increased risk of hypertension and cardiovascular disease on the offspring and the postpartum mother later in life. The exact cause of preeclampsia is unknown, but women with preeclampsia have elevated concentrations of agonistic autoantibodies against the angiotensin II type 1 receptor (AT1-AA). These AT1-AA's through multiple studies have shown to play a significant role in the pathology and possible genesis of preeclampsia. This review will discuss the discovery of AT1-AAs and the role of AT1-AAs in the pathophysiology of preeclampsia. This review will also discuss future therapeutic approaches towards the AT1-AA to prevent adverse pregnancy outcomes. Furthermore, we will examine the relationship between AT1-AA induced hypertension associated with increased oxidative stress, antiangiogenic factors (such as soluble fms-related tyrosine kinase-1 (sFlt-1), endothelin-1 (ET-1), inflammation, endothelial dysfunction, and reduced renal function. Understanding the pathological role of AT1-AAs in hypertensive pregnancies is important as we search for novel therapies to manage preeclampsia.
Collapse
Affiliation(s)
- Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - Mark W Cunningham
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
154
|
Cottrell JN, Amaral LM, Harmon A, Cornelius DC, Cunningham MW, Vaka VR, Ibrahim T, Herse F, Wallukat G, Dechend R, LaMarca B. Interleukin-4 supplementation improves the pathophysiology of hypertension in response to placental ischemia in RUPP rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R165-R171. [PMID: 30624978 DOI: 10.1152/ajpregu.00167.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia (PE) is characterized by chronic inflammation and elevated agonistic autoantibodies to the angiotensin type 1 receptor (AT1-AA), endothelin-1, and uterine artery resistance index (UARI) during pregnancy. Previous studies report an imbalance among immune cells, with T-helper type 2 (Th2) cells being decreased during PE. We hypothesized that interleukin-4 (IL-4) would increase Th2 cells and improve the pathophysiology in response to placental ischemia during pregnancy. IL-4 (600 ng/day) was administered via osmotic minipump on gestational day 14 to normal pregnant (NP) and reduced uterine perfusion pressure (RUPP) rats. Carotid catheters were inserted, and Doppler ultrasound was performed on gestational day 18. Blood pressure (mean arterial pressure), TNF-α, IL-6, AT1-AA, natural killer cells, Th2 cells, and B cells were measured on gestational day 19. Mean arterial pressure was 97 ± 2 mmHg in NP ( n = 9), 101 ± 3 mmHg in IL-4-treated NP ( n = 14), and 137 ± 4 mmHg in RUPP ( n = 8) rats and improved to 108 ± 3 mmHg in IL-4-treated RUPP rats ( n = 17) ( P < 0.05). UARI was 0.5 ± 0.03 in NP and 0.8 in RUPP rats and normalized to 0.5 in IL-4-treated RUPP rats ( P < 0.05). Plasma nitrate-nitrite levels increased in IL-4-treated RUPP rats, while placental preproendothelin-1 expression, plasma TNF-α and IL-6, and AT1-AA decreased in IL-4-treated RUPP rats compared with untreated RUPP rats ( P < 0.05). Circulating B cells and placental cytolytic natural killer cells decreased after IL-4 administration, while Th2 cells increased in IL-4-treated RUPP compared with untreated RUPP rats. This study illustrates that IL-4 decreased inflammation and improved Th2 numbers in RUPP rats and, ultimately, improved hypertension in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Jesse N Cottrell
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Ashlyn Harmon
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Mark W Cunningham
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Venkata Ramana Vaka
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Florian Herse
- HELIOS Clinic, Charité, Campus-Buch and Max-Delbrueck Center , Berlin , Germany
| | - Gerd Wallukat
- HELIOS Clinic, Charité, Campus-Buch and Max-Delbrueck Center , Berlin , Germany
| | - Ralf Dechend
- HELIOS Clinic, Charité, Campus-Buch and Max-Delbrueck Center , Berlin , Germany
| | - Babbette LaMarca
- Department of Pharmacology, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
155
|
|
156
|
Skalis G, Katsi V, Miliou A, Georgiopoulos G, Papazachou O, Vamvakou G, Nihoyannopoulos P, Tousoulis D, Makris T. MicroRNAs in Preeclampsia. Microrna 2019; 8:28-35. [PMID: 30101723 DOI: 10.2174/2211536607666180813123303] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 05/22/2018] [Accepted: 07/27/2018] [Indexed: 11/22/2022]
Abstract
Preeclampsia (PE) continues to represent a worldwide problem and challenge for both clinicians and laboratory-based doctors. Despite many efforts, the knowledge acquired regarding its pathogenesis and pathophysiology does not allow us to treat it efficiently. It is not possible to arrest its progressive nature, and the available therapies are limited to symptomatic treatment. Furthermore, both the diagnosis and prognosis are frequently uncertain, whilst the ability to predict its occurrence is very limited. MicroRNAs are small non-coding RNAs discovered two decades ago, and present great interest given their ability to regulate almost every aspect of the cell function. A lot of evidence regarding the role of miRNAs in pre-eclampsia has been accumulated in the last 10 years. Differentially expressed miRNAs are characteristic of both mild and severe PE. In many cases they target signaling pathway-related genes that result in altered processes which are directly involved in PE. Immune system, angiogenesis and trophoblast proliferation and invasion, all fundamental aspects of placentation, are controlled in various degrees by miRNAs which are up- or downregulated. Finally, miRNAs represent a potential therapeutic target and a diagnostic tool.
Collapse
Affiliation(s)
- Georgios Skalis
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| | - Vasiliki Katsi
- Cardiology Department, Hippokration Hospital, National Health System, Athens, Greece
| | - Antigoni Miliou
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | | | | | - Georgia Vamvakou
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| | - Petros Nihoyannopoulos
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Thomas Makris
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| |
Collapse
|
157
|
Lumbers ER, Delforce SJ, Arthurs AL, Pringle KG. Causes and Consequences of the Dysregulated Maternal Renin-Angiotensin System in Preeclampsia. Front Endocrinol (Lausanne) 2019; 10:563. [PMID: 31551925 PMCID: PMC6746881 DOI: 10.3389/fendo.2019.00563] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022] Open
Abstract
A healthy pregnancy outcome depends on the activation of the renin-angiotensin-aldosterone system (RAAS) as a regulated, integrated response to the growing demands of the conceptus. Both the circulating RAAS and the intrarenal renin-angiotensin system (iRAS) play major roles in cardiovascular function and fluid and electrolyte homeostasis. The circulating RAAS becomes dysfunctional in preeclampsia and we propose that dysregulation of the iRAS plays a role in development of the clinical syndrome known as preeclampsia. Experimental studies in animals have shown that placental renin, when released into the maternal circulation, can cause hypertension. We postulate that abnormal placental development is associated with over-secretion of renin and other RAS proteins/angiotensin (Ang) peptides by the placenta/decidua into the maternal circulation. We hypothesise that this is because of increased shedding of exosomes and other placental particles into the maternal circulation that not only contain RAS proteins and peptides but also microRNAs (miRNAs) that target RAS mRNAs, and Ang II type 1 receptor autoantibodies (AT1R-AAs), that are agonists for, and have the same actions as, Ang II. As a result, there is both suppression of the circulating RAAS that is responsible for maintaining maternal homeostasis and activation of the iRAS. Together with altered vascular reactivity to Ang peptides, the iRAS causes hypertension, renal damage and secondary changes in the neurohumoral control of the maternal circulation and fluid and electrolyte balance, which contribute to the pathophysiology of preeclampsia.
Collapse
Affiliation(s)
- Eugenie R. Lumbers
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle upon Tyne, NSW, Australia
- *Correspondence: Eugenie R. Lumbers
| | - Sarah J. Delforce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle upon Tyne, NSW, Australia
| | - Anya L. Arthurs
- Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, SA, Australia
| | - Kirsty G. Pringle
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Priority Research Centre for Reproductive Sciences, University of Newcastle, Newcastle upon Tyne, NSW, Australia
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle upon Tyne, NSW, Australia
| |
Collapse
|
158
|
Sabbadin C, Andrisani A, Ambrosini G, Bordin L, Donà G, Manso J, Ceccato F, Scaroni C, Armanini D. Aldosterone in Gynecology and Its Involvement on the Risk of Hypertension in Pregnancy. Front Endocrinol (Lausanne) 2019; 10:575. [PMID: 31507531 PMCID: PMC6716345 DOI: 10.3389/fendo.2019.00575] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
Aldosterone is the main mineralocorticoid hormone, responsible of the regulation of fluid and electrolyte balance and blood pressure. It acts also as a pro-inflammatory factor responsible of an increased cardiovascular risk, independent from blood pressure values. After the discovery of mineralocorticoid receptor (MR) in mononuclear leukocytes, further studies supported its role in inflammatory and even autoimmune mechanisms underlying several diseases. In particular, recent studies reported a possible involvement of aldosterone in some gynecological conditions and diseases, characterized by inflammation, hypertension and increased cardio-metabolic risk, such as use of hormonal contraceptives, preeclampsia, polycystic ovary syndrome, uterine fibroids, and endometriosis. The aim of this mini-review is to report the possible involvement of aldosterone in all these gynecological conditions, suggesting different pathogenetic mechanisms and new target treatments of MR blockers for these diseases.
Collapse
Affiliation(s)
- Chiara Sabbadin
- Department of Medicine–Endocrinology, University of Padova, Padua, Italy
| | | | - Guido Ambrosini
- Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Luciana Bordin
- Department of Molecular Medicine-Biological Chemistry, University of Padova, Padua, Italy
| | - Gabriella Donà
- Department of Molecular Medicine-Biological Chemistry, University of Padova, Padua, Italy
| | - Jacopo Manso
- Department of Medicine–Endocrinology, University of Padova, Padua, Italy
| | - Filippo Ceccato
- Department of Medicine–Endocrinology, University of Padova, Padua, Italy
| | - Carla Scaroni
- Department of Medicine–Endocrinology, University of Padova, Padua, Italy
| | - Decio Armanini
- Department of Medicine–Endocrinology, University of Padova, Padua, Italy
- *Correspondence: Decio Armanini
| |
Collapse
|
159
|
Marshall SA, Cox AG, Parry LJ, Wallace EM. Targeting the vascular dysfunction: Potential treatments for preeclampsia. Microcirculation 2018; 26:e12522. [PMID: 30556222 DOI: 10.1111/micc.12522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/22/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a pregnancy-specific disorder, primarily characterized by new-onset hypertension in combination with a variety of other maternal or fetal signs. The pathophysiological mechanisms underlying the disease are still not entirely clear. Systemic maternal vascular dysfunction underlies the clinical features of preeclampsia. It is a result of oxidative stress and the actions of excessive anti-angiogenic factors, such as soluble fms-like tyrosine kinase, soluble endoglin, and activin A, released by a dysfunctional placenta. The vascular dysfunction then leads to impaired regulation and secretion of relaxation factors and an increase in sensitivity/production of constrictors. This results in a more constricted vasculature rather than the relaxed vasodilated state associated with normal pregnancy. Currently, the only effective "treatment" for preeclampsia is delivery of the placenta and therefore the baby. Often, this means a preterm delivery to save the life of the mother, with all the attendant risks and burdens associated with fetal prematurity. To lessen this burden, there is a pressing need for more effective treatments that target the maternal vascular dysfunction that underlies the hypertension. This review details the vascular effects of key drugs undergoing clinical assessment as potential treatments for women with preeclampsia.
Collapse
Affiliation(s)
- Sarah A Marshall
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| | - Annie G Cox
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| | - Laura J Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Euan M Wallace
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
160
|
Cunningham MW, Vaka VR, McMaster K, Ibrahim T, Cornelius DC, Amaral L, Campbell N, Wallukat G, McDuffy S, Usry N, Dechend R, LaMarca B. Renal natural killer cell activation and mitochondrial oxidative stress; new mechanisms in AT1-AA mediated hypertensive pregnancy. Pregnancy Hypertens 2018; 15:72-77. [PMID: 30825931 DOI: 10.1016/j.preghy.2018.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022]
Abstract
Women with preeclampsia (PE) have increased mean arterial pressure (MAP), natural killer (NK) cells, reactive oxygen species (ROS), and agonistic autoantibodies to the angiotensin II type 1 receptor (AT1-AA). AT1-AA's administered to pregnant rodents produces a well-accepted model of PE. However, the role of NK cells and mitochondrial reactive oxygen species (mtROS) in AT1-AA mediated hypertension during pregnancy is unknown. We hypothesize that AT1-AA induced model of PE will exhibit elevated MAP, NK cells, and mtROS; while inhibition of the AT1-AA binding to the AT1R would be preventative. Pregnant rats were divided into 4 groups: normal pregnant (NP) (n = 5), NP + AT1-AA inhibitory peptide (NP +'n7AAc') (n = 3), NP + AT1-AA infused (NP + AT1-AA) (n = 10), and NP + AT1-AA +'n7AAc' (n = 8). Day 13, rats were surgically implanted with mini-pumps infusing either AT1-AA or AT1-AA +'n7AAc'. Day 19, tissue and blood was collected. MAP was elevated in AT1-AA vs. NP (119 ± 1 vs. 102 ± 2 mmHg, p < 0.05) and this was prevented by 'n7AAc' (108 ± 3). There was a 6 fold increase in renal activated NK cells in AT1-AA vs NP (1.2 ± 0.4 vs. 0.2 ± 0.1% Gated, p = 0.05) which returned to NP levels in AT1-AA +'n7AAc' (0.1 ± 0.1% Gated). Renal mtROS (317 ± 49 vs. 101 ± 13% Fold, p < 0.05) was elevated with AT1-AA vs NP and was decreased in AT1-AA +'n7AAc' (128 ± 16, p < 0.05). In conclusion, AT1-AA's increased MAP, NK cells, and mtROS which were attenuated by AT1-AA inhibition, thus highlighting new mechanisms of AT1-AA and the importance of drug therapy targeted to AT1-AAs in hypertensive pregnancies.
Collapse
Affiliation(s)
- Mark W Cunningham
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Venkata Ramana Vaka
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kristen McMaster
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Tarek Ibrahim
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Denise C Cornelius
- Depart. of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lorena Amaral
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Nathan Campbell
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Gerd Wallukat
- Experimental and Clinical Research Center, Charité Campus Buch, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Shyanne McDuffy
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Nathan Usry
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Berlin, Germany
| | - Babbette LaMarca
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States; Depart. of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States.
| |
Collapse
|
161
|
Quitterer U, Fu X, Pohl A, Bayoumy KM, Langer A, AbdAlla S. Beta-Arrestin1 Prevents Preeclampsia by Downregulation of Mechanosensitive AT1-B2 Receptor Heteromers. Cell 2018; 176:318-333.e19. [PMID: 30503206 DOI: 10.1016/j.cell.2018.10.050] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/26/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
Preeclampsia is the most frequent pregnancy-related complication worldwide with no cure. While a number of molecular features have emerged, the underlying causal mechanisms behind the disorder remain obscure. Here, we find that increased complex formation between angiotensin II AT1 and bradykinin B2, two G protein-coupled receptors with opposing effects on blood vessel constriction, triggers symptoms of preeclampsia in pregnant mice. Aberrant heteromerization of AT1-B2 led to exaggerated calcium signaling and high vascular smooth muscle mechanosensitivity, which could explain the onset of preeclampsia symptoms at late-stage pregnancy as mechanical forces increase with fetal mass. AT1-B2 receptor aggregation was inhibited by beta-arrestin-mediated downregulation. Importantly, symptoms of preeclampsia were prevented by transgenic ARRB1 expression or a small-molecule drug. Because AT1-B2 heteromerization was found to occur in human placental biopsies from pregnancies complicated by preeclampsia, specifically targeting AT1-B2 heteromerization and its downstream consequences represents a promising therapeutic approach.
Collapse
Affiliation(s)
- Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Department of Medicine, Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Xuebin Fu
- Department of Microbiology and Immunology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Armin Pohl
- Roche Diagnostics International AG, Forrenstrasse 2, 6343 Rotkreuz, Switzerland
| | - Karam M Bayoumy
- Clinic of Obstetrics and Gynecology, Ain Shams University Hospitals, Cairo 11566, Egypt
| | - Andreas Langer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Said AbdAlla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
162
|
Bussolino S, Dolla C, Ariaudo C, Civiletti F, Messina M, Mella A, Caorsi C, Amoroso A, Barreca A, Papotti M, Giunti S, Fop F, Biancone L. Detection of Angiotensin II type I-receptor antibodies in transplant glomerulopathy. Clin Transplant 2018; 32:e13407. [DOI: 10.1111/ctr.13407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 08/28/2018] [Accepted: 09/08/2018] [Indexed: 01/18/2023]
Affiliation(s)
| | - Caterina Dolla
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Claudia Ariaudo
- Division of Nephrology and Dialysis; ASO S. Croce e Carle; Cuneo Italy
| | - Federica Civiletti
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Maria Messina
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Alberto Mella
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Cristiana Caorsi
- Immunogenetic and Transplant Biology Center, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Antonio Amoroso
- Immunogenetic and Transplant Biology Center, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Antonella Barreca
- Pathology Division, Department of Oncology; University of Turin; Turin Italy
| | - Mauro Papotti
- Pathology Division, Department of Oncology; University of Turin; Turin Italy
| | - Sara Giunti
- Department of Medical Sciences; University of Turin; Turin Italy
| | - Fabrizio Fop
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| | - Luigi Biancone
- Renal Transplantation Center, “A. Vercellone”, Division of Nephrology Dialysis and Transplantation, Città della Salute e della Scienza Hospital and Department of Medical Sciences; University of Turin; Turin Italy
| |
Collapse
|
163
|
Maher GM, McCarthy FP, McCarthy CM, Kenny LC, Kearney PM, Khashan AS, O'Keeffe GW. A perspective on pre-eclampsia and neurodevelopmental outcomes in the offspring: Does maternal inflammation play a role? Int J Dev Neurosci 2018; 77:69-76. [PMID: 30391740 DOI: 10.1016/j.ijdevneu.2018.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 10/28/2022] Open
Abstract
Pre-eclampsia is a leading cause of maternal death and maternal and perinatal morbidity. Whilst the clinical manifestations of pre-eclampsia often occur in late pregnancy, the molecular events leading into the onset of this disease are thought to originate in early pregnancy and result in insufficient placentation. Although the causative molecular basis of pre-eclampsia remains poorly understood, maternal inflammation is recognised as a core clinical feature. While the adverse effects of pre-eclampsia on maternal and fetal health in pregnancy is well-recognised, the long-term impact of pre-eclampsia exposure on the risk of autism spectrum disorder (ASD) in exposed offspring is a topic of on-going debate. In particular, a recent systematic review has reported an association between exposure to pre-eclampsia and increased risk of ASD, however the molecular basis of this association is unknown. Here we review recent evidence for; 1) maternal inflammation in pre-eclampsia; 2) epidemiological evidence for alterations in neurodevelopmental outcomes in offspring exposed to pre-eclampsia; 3) long-term changes in the brains of offspring exposed to pre-eclampsia; and 4) how maternal inflammation may lead to altered neurodevelopmental outcomes in pre-eclampsia exposed offspring. Finally, we discuss the implications of this for the development of future studies in this field.
Collapse
Affiliation(s)
- Gillian M Maher
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland.,The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland
| | - Fergus P McCarthy
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland.,Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, Ireland
| | - Cathal M McCarthy
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland.,Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, United Kingdom
| | - Patricia M Kearney
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland
| | - Ali S Khashan
- School of Public Health, Western Gateway Building, University College Cork, Cork, Ireland.,The Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital and University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
164
|
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
|
165
|
Parchem JG, Kanasaki K, Kanasaki M, Sugimoto H, Xie L, Hamano Y, Lee SB, Gattone VH, Parry S, Strauss JF, Garovic VD, McElrath TF, Lu KH, Sibai BM, LeBleu VS, Carmeliet P, Kalluri R. Loss of placental growth factor ameliorates maternal hypertension and preeclampsia in mice. J Clin Invest 2018; 128:5008-5017. [PMID: 30179860 PMCID: PMC6205389 DOI: 10.1172/jci99026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/28/2018] [Indexed: 12/28/2022] Open
Abstract
Preeclampsia remains a clinical challenge due to its poorly understood pathogenesis. A prevailing notion is that increased placental production of soluble fms-like tyrosine kinase-1 (sFlt-1) causes the maternal syndrome by inhibiting proangiogenic placental growth factor (PlGF) and VEGF. However, the significance of PlGF suppression in preeclampsia is uncertain. To test whether preeclampsia results from the imbalance of angiogenic factors reflected by an abnormal sFlt-1/PlGF ratio, we studied PlGF KO (Pgf-/-) mice and noted that the mice did not develop signs or sequelae of preeclampsia despite a marked elevation in circulating sFLT-1. Notably, PlGF KO mice had morphologically distinct placentas, showing an accumulation of junctional zone glycogen. We next considered the role of placental PlGF in an established model of preeclampsia (pregnant catechol-O-methyltransferase-deficient [COMT-deficient] mice) by generating mice with deletions in both the Pgf and Comt genes. Deletion of placental PlGF in the context of COMT loss resulted in a reduction in maternal blood pressure and increased placental glycogen, indicating that loss of PlGF might be protective against the development of preeclampsia. These results identify a role for PlGF in placental development and support a complex model for the pathogenesis of preeclampsia beyond an angiogenic factor imbalance.
Collapse
Affiliation(s)
- Jacqueline G Parchem
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Megumi Kanasaki
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Liang Xie
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuki Hamano
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Soo Bong Lee
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Vincent H Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samuel Parry
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Vesna D Garovic
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas F McElrath
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Karen H Lu
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
166
|
Yu W, Gao W, Rong D, Wu Z, Khalil RA. Molecular determinants of microvascular dysfunction in hypertensive pregnancy and preeclampsia. Microcirculation 2018; 26:e12508. [PMID: 30338879 PMCID: PMC6474836 DOI: 10.1111/micc.12508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022]
Abstract
Preeclampsia is a pregnancy-related disorder characterized by hypertension and often fetal intrauterine growth restriction, but the underlying mechanisms are unclear. Defective placentation and apoptosis of invasive cytotrophoblasts cause inadequate remodeling of spiral arteries, placental ischemia, and reduced uterine perfusion pressure (RUPP). RUPP causes imbalance between the anti-angiogenic factors soluble fms-like tyrosine kinase-1 and soluble endoglin and the pro-angiogenic vascular endothelial growth factor and placental growth factor, and stimulates the release of proinflammatory cytokines, hypoxia-inducible factor, reactive oxygen species, and angiotensin AT1 receptor agonistic autoantibodies. These circulating factors target the vascular endothelium, smooth muscle and various components of the extracellular matrix. Generalized endotheliosis in systemic, renal, cerebral, and hepatic vessels causes decreases in endothelium-derived vasodilators such as nitric oxide, prostacyclin and hyperpolarization factor, and increases in vasoconstrictors such as endothelin-1 and thromboxane A2. Enhanced mechanisms of vascular smooth muscle contraction, such as intracellular Ca2+ , protein kinase C, and Rho-kinase cause further increases in vasoconstriction. Changes in matrix metalloproteinases and extracellular matrix cause inadequate vascular remodeling and increased arterial stiffening, leading to further increases in vascular resistance and hypertension. Therapeutic options are currently limited, but understanding the molecular determinants of microvascular dysfunction could help in the design of new approaches for the prediction and management of preeclampsia.
Collapse
Affiliation(s)
- Wentao Yu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wei Gao
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dan Rong
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhixian Wu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
167
|
Buttrup Larsen S, Wallukat G, Schimke I, Sandager A, Tvilum Christensen T, Uldbjerg N, Tørring N. Functional autoantibodies against Endothelin-1 receptor type A and Angiotensin II receptor type 1 in patients with preeclampsia. Pregnancy Hypertens 2018; 14:189-194. [DOI: 10.1016/j.preghy.2018.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/05/2018] [Accepted: 10/12/2018] [Indexed: 12/25/2022]
|
168
|
Eddy AC, Chapman H, George EM. Acute Hypoxia and Chronic Ischemia Induce Differential Total Changes in Placental Epigenetic Modifications. Reprod Sci 2018; 26:766-773. [PMID: 30223723 DOI: 10.1177/1933719118799193] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Preeclampsia is a common obstetrical complication, hallmarked by new-onset hypertension. Believed to result from placental insufficiency and chronic placental ischemia, the symptoms of preeclampsia are caused by release of pathogenic factors from the placenta itself, although the mechanisms of their regulation are in many cases unknown. One potential mechanism is through changes in placental epigenetic chromatin modifications, particularly histone acetylation and DNA methylation. Here, we determined the effects of chronic ischemia on global epigenetic modifications in the rodent placenta in vivo and acute hypoxia in BeWo placental trophoblast cells in vitro. Placental insufficiency via uterine artery restriction increased maternal blood pressure and fetal demise while decreasing placental and fetal mass. Global placental histone H3 acetylation levels were significantly decreased at H3 K9, K14, K18, K27, and K56. Interestingly, when BeWo-immortalized placental trophoblast cells were cultured in oxygen concentrations mimicking healthy and ischemic placentas, there was a significant increase in acetylated at K9, K18, K27, and K56. This was associated with a small but significant decrease in placental acetyl-CoA, suggesting depletion in the source of acetyl group donors. Finally, while global methylation of cytosine from placental DNA was low in both groups of animals (<1%), there was ∼50% increase in 5-mC in response to chronic ischemia. This suggests acute hypoxia and chronic ischemia induce differential global changes in histone acetylation in the placenta and that chronically altered metabolic profiles could affect histone acetylation in the placenta, thereby regulating production of pathogenic factors from the placenta during preeclampsia.
Collapse
Affiliation(s)
- Adrian C Eddy
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | - Heather Chapman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA. .,Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
169
|
Stanhewicz AE. Residual vascular dysfunction in women with a history of preeclampsia. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1062-R1071. [PMID: 30133302 DOI: 10.1152/ajpregu.00204.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy characterized by new-onset hypertension, proteinuria, and edema occurring after 20 wk of gestation, with a prevalence of ~7-10% of pregnancies in the United States and ~8 million pregnancies worldwide. Despite the postpartum remission of preeclamptic symptoms, women who have had preeclampsia are two to four times more likely to develop cardiovascular disease (CVD) and are significantly more likely to die of CVD compared with women with a history of normal pregnancy. Although the relation between history of preeclampsia and elevated CVD risk is well documented, the mechanism(s) underlying this association remains unclear. One hypothesis explaining this association is that the initial vascular damage and dysfunction sustained during the preeclamptic pregnancy persist chronically. Indeed, even in the absence of, or in advance of, overt CVD women who have had preeclampsia have compromised vascular endothelial function. Emerging mechanistic studies in these women have provided some insight into the underlying mechanisms of this persistent vascular dysfunction and have begun to identify potential therapeutic targets for the prevention or mitigation of CVD progression in this vulnerable population. This review summarizes the existing literature examining vascular function and dysfunction in women with a history of preeclampsia and highlights future directions for mechanistic investigations and development of novel intervention strategies aimed at halting or slowing the progression of CVD in these women.
Collapse
Affiliation(s)
- Anna E Stanhewicz
- Department of Kinesiology, Pennsylvania State University , University Park, Pennsylvania
| |
Collapse
|
170
|
Lu J, Wang Z, Cao J, Chen Y, Dong Y. A novel and compact review on the role of oxidative stress in female reproduction. Reprod Biol Endocrinol 2018; 16:80. [PMID: 30126412 PMCID: PMC6102891 DOI: 10.1186/s12958-018-0391-5] [Citation(s) in RCA: 300] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/23/2018] [Indexed: 12/30/2022] Open
Abstract
In recent years, the study of oxidative stress (OS) has become increasingly popular. In particular, the role of OS on female fertility is very important and has been focused on closely. The occurrence of OS is due to the excessive production of reactive oxygen species (ROS). ROS are a double-edged sword; they not only play an important role as secondary messengers in many intracellular signaling cascades, but they also exert indispensable effects on pathological processes involving the female genital tract. ROS and antioxidants join in the regulation of reproductive processes in both animals and humans. Imbalances between pro-oxidants and antioxidants could lead to a number of female reproductive diseases. This review focuses on the mechanism of OS and a series of female reproductive processes, explaining the role of OS in female reproduction and female reproductive diseases caused by OS, including polycystic ovary syndrome (PCOS), endometriosis, preeclampsia and so on. Many signaling pathways involved in female reproduction, including the Keap1-Nrf2, NF-κB, FOXO and MAPK pathways, which are affected by OS, are described, providing new ideas for the mechanism of reproductive diseases.
Collapse
Affiliation(s)
- Jiayin Lu
- Laboratory of Neurobiology, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193 People’s Republic of China
| | - Zixu Wang
- Laboratory of Neurobiology, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193 People’s Republic of China
| | - Jing Cao
- Laboratory of Neurobiology, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193 People’s Republic of China
| | - Yaoxing Chen
- Laboratory of Neurobiology, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193 People’s Republic of China
| | - Yulan Dong
- Laboratory of Neurobiology, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193 People’s Republic of China
| |
Collapse
|
171
|
Sabbadin C, Ceccato F, Ragazzi E, Boscaro M, Betterle C, Armanini D. Evaluation of angiotensin II type-1 receptor antibodies in primary aldosteronism and further considerations about their possible pathogenetic role. J Clin Hypertens (Greenwich) 2018; 20:1313-1318. [PMID: 30058103 DOI: 10.1111/jch.13351] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/09/2018] [Accepted: 06/16/2018] [Indexed: 12/18/2022]
Abstract
Angiotensin II type-1 receptor autoantibodies (AT1RAb) have been involved in the genesis of primary aldosteronism (PA), both in aldosterone-producing adenoma (APA) and in idiopathic hyperaldosteronism (IHA). In this study, we evaluated the titer of AT1RAb in 44 PA patients (15 with APA and 29 with IHA) compared with 18 normotensive healthy controls who were matched for gender and age. In 17 PA patients (6 APA and 11 IHA) the titer was evaluated under mineralocorticoid receptor (MR) antagonist treatment. We found that PA patients had a significantly higher titer of AT1RAb compared with controls (median values 33 [IQR 15.6] IU/mL vs 17.5 [IQR 10.8] IU/mL, respectively; P < 0.0001). No significant difference of the AT1RAb titer was reported among PA patients, subdivided according to the subtypes and the concomitant MR antagonist therapy. No significant correlation was detected between age, gender, BMI, blood pressure values, baseline aldosterone, ARR, and the AT1RAb titer of all patients enrolled. Our data confirm an increased titer of AT1RAb in both subtypes of PA, independently from the concomitant use of MR antagonists and clinical/biochemical characteristics of PA patients. The small sample of patients and the relatively short time of treatment could have influenced these results. Moreover, the ELISA assay fails to evaluate the bioactivity of AT1RAb. Further studies should evaluate if the subtype, the clinical/biochemical recovery of PA, or both, influence the pathogenetic role of AT1RAb. The possible autoimmune pathogenesis and reversal effect with AT1R blocker treatment in PA patients with AT1RAb positivity is intriguing and requires further study.
Collapse
Affiliation(s)
- Chiara Sabbadin
- Department of Medicine, Endocrinology, University of Padova, Padova, Italy
| | - Filippo Ceccato
- Department of Medicine, Endocrinology, University of Padova, Padova, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Marco Boscaro
- Department of Medicine, Endocrinology, University of Padova, Padova, Italy
| | - Corrado Betterle
- Department of Medicine, Endocrinology, University of Padova, Padova, Italy
| | - Decio Armanini
- Department of Medicine, Endocrinology, University of Padova, Padova, Italy
| |
Collapse
|
172
|
Liu Z, Khalil RA. Evolving mechanisms of vascular smooth muscle contraction highlight key targets in vascular disease. Biochem Pharmacol 2018; 153:91-122. [PMID: 29452094 PMCID: PMC5959760 DOI: 10.1016/j.bcp.2018.02.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/12/2018] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle (VSM) plays an important role in the regulation of vascular function. Identifying the mechanisms of VSM contraction has been a major research goal in order to determine the causes of vascular dysfunction and exaggerated vasoconstriction in vascular disease. Major discoveries over several decades have helped to better understand the mechanisms of VSM contraction. Ca2+ has been established as a major regulator of VSM contraction, and its sources, cytosolic levels, homeostatic mechanisms and subcellular distribution have been defined. Biochemical studies have also suggested that stimulation of Gq protein-coupled membrane receptors activates phospholipase C and promotes the hydrolysis of membrane phospholipids into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 stimulates initial Ca2+ release from the sarcoplasmic reticulum, and is buttressed by Ca2+ influx through voltage-dependent, receptor-operated, transient receptor potential and store-operated channels. In order to prevent large increases in cytosolic Ca2+ concentration ([Ca2+]c), Ca2+ removal mechanisms promote Ca2+ extrusion via the plasmalemmal Ca2+ pump and Na+/Ca2+ exchanger, and Ca2+ uptake by the sarcoplasmic reticulum and mitochondria, and the coordinated activities of these Ca2+ handling mechanisms help to create subplasmalemmal Ca2+ domains. Threshold increases in [Ca2+]c form a Ca2+-calmodulin complex, which activates myosin light chain (MLC) kinase, and causes MLC phosphorylation, actin-myosin interaction, and VSM contraction. Dissociations in the relationships between [Ca2+]c, MLC phosphorylation, and force have suggested additional Ca2+ sensitization mechanisms. DAG activates protein kinase C (PKC) isoforms, which directly or indirectly via mitogen-activated protein kinase phosphorylate the actin-binding proteins calponin and caldesmon and thereby enhance the myofilaments force sensitivity to Ca2+. PKC-mediated phosphorylation of PKC-potentiated phosphatase inhibitor protein-17 (CPI-17), and RhoA-mediated activation of Rho-kinase (ROCK) inhibit MLC phosphatase and in turn increase MLC phosphorylation and VSM contraction. Abnormalities in the Ca2+ handling mechanisms and PKC and ROCK activity have been associated with vascular dysfunction in multiple vascular disorders. Modulators of [Ca2+]c, PKC and ROCK activity could be useful in mitigating the increased vasoconstriction associated with vascular disease.
Collapse
Affiliation(s)
- Zhongwei Liu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
173
|
Wang P, Zhang S, Ren J, Yan L, Bai L, Wang L, Wang P, Bian J, Yin X, Liu H. The inhibitory effect of BKCa channels induced by autoantibodies against angiotensin II type 1 receptor is independent of AT1R. Acta Biochim Biophys Sin (Shanghai) 2018; 50:560-566. [PMID: 29697782 DOI: 10.1093/abbs/gmy038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 12/28/2022] Open
Abstract
Autoantibodies against angiotensin II Type 1 receptor (AT1-AA) are routinely detected in the serum of preeclampsia patients, which results in an increase in vascular tone and an elevation in intracellular calcium concentration of rat vascular smooth muscle (VSM). The big conductance calcium-activated potassium channels (BKCa channels) account for the dominant outward currents in VSMCs, contributing to membrane hyperpolarization and vasodilation. In the present study, we investigated the effect of AT1-AA on BKCa channels. A preeclampsia model was established by passively immunizing healthy pregnant BALB/c mice with AT1-AA extracted from hybridoma culture supernatant. Blood pressure, serum AT1-AA levels, and urinary protein were measured in the immunized mice. BKCa channel expression was detected using qRT-PCR and immunohistochemical technique. The patch-clamp technique was used to record the single currents of BKCa channels in the HEK293T cells that had been transfected. AT1-AA immunized mice exhibited elevated AT1-AA and urinary protein levels compared with mice of the vehicle group. Systolic blood pressure was also increased in the immunized group. BKCa channel β1-subunit expression was reduced in the mesenteric arteries of immunized mice. AT1-AA could inhibit the BKCa currents and the inhibitory effects were not completely reversed following the application of valsartan, an inhibitor of AT1 receptor. In conclusion, AT1-AA could decrease BKCa expression and inhibit BKCa activity independent of AT1R. These inhibitory effects are likely to be contributory factors in the promotion of increased vascular tone caused by AT1-AA in preeclampsia.
Collapse
Affiliation(s)
- Peng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jie Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100069, China
| | - Lina Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Pengli Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jingwei Bian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaochen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing 100069, China
| |
Collapse
|
174
|
Prevalence of Antibody against Angiotensin II Type 1 Receptor (AT1R) Among Thai Kidney Transplant Patients. Transplant Proc 2018; 50:1310-1315. [DOI: 10.1016/j.transproceed.2018.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
|
175
|
Angiotensin II responsiveness after preeclampsia: translational data from an experimental rat model and early-onset human preeclampsia. J Hypertens 2018; 35:2468-2478. [PMID: 28708773 DOI: 10.1097/hjh.0000000000001474] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Formerly preeclamptic women have an increased risk for cardiovascular and renal disease later in life. It is unknown which mechanisms contribute to this increased risk and whether this is induced by preeclampsia or by prepregnancy factors. We hypothesized that the increased risk for cardiovascular disease is partly due to an increased angiotensin II (ang II) responsiveness postpartum and that preeclampsia itself is involved in inducing this increased ang II responsiveness. METHODS In never-pregnant, formerly healthy pregnant rats and rats with former experimental preeclampsia [experimental preeclampsia model induced by low-dose endotoxin infusion on day 14 of pregnancy; endotoxin-infused pregnant rats (EP-rats)], ang II responsiveness was studied by measuring changes in blood pressure (BP) and proteinuria after chronic ang II infusion with osmotic minipumps (200 ng/kg per min). In addition, we measured BP and responses to ang II (0.3, 1.0 and 3.0 ng/kg per min) in 18 formerly early-onset preeclamptic, without comorbidities, and 18 formerly healthy pregnant women (controls). RESULTS In rats, a significantly higher systolic BP at termination was observed in formerly EP-rats vs. never-pregnant rats after ang II infusion (159.5 ± 29.5 vs. 136.7 ± 16.8; P = 0.049). In response to ang II, there was a significant increase in proteinuria in formerly EP-rats vs. healthy pregnant and never-pregnant rats (P < 0.01 for both). In humans, 1.0 ng/kg per min ang II showed a trend towards an increased mean arterial BP response in formerly preeclamptic women vs. controls (P = 0.057). CONCLUSION Our data show an increased ang II responsiveness following (experimental) preeclampsia and support a role for preeclampsia itself in altered ang II responsiveness postpartum.
Collapse
|
176
|
Auto-antibodies against the angiotensin II type I receptor in women with uteroplacental acute atherosis and preeclampsia at delivery and several years postpartum. J Reprod Immunol 2018; 128:23-29. [PMID: 29843114 DOI: 10.1016/j.jri.2018.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/16/2018] [Accepted: 05/21/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Uteroplacental acute atherosis is a pregnancy-specific lesion resembling early stages of atherosclerosis found frequently in preeclampsia. Preeclampsia is associated with an increased risk for future maternal atherosclerotic cardiovascular disease. The renin-angiotensin-system plays a role both in atherosclerosis and in preeclampsia. Circulating agonistic autoantibodies at the angiotensin-II type 1 receptor (AT1-AA) are increased in preeclampsia. We hypothesized an association between AT1-AA at delivery and postpartum with acute atherosis in pregnancy. MATERIAL AND METHODS Maternal serum and decidua basalis tissue was collected at elective cesarean section (n = 41; 24 preeclampsia, 17 normotensive controls). Circulating AT1-AA were detected by a bioassay using spontaneously beating rat cardiomyocytes at delivery (n = 41) and 5-8 years postpartum in a subgroup (n = 10). Decidual acute atherosis was assessed by immunohistochemistry. RESULTS Significantly less normotensive controls (18%; 3/17) than women with preeclampsia (58%; 14/24) were AT1-AA positive at delivery, p<0.01. Uteroplacental acute atherosis and circulating AT1-AA at delivery were not significantly correlated. Postpartum, 2 prior preeclamptic women had circulating AT1-AA, both without acute atherosis in pregnancy. CONCLUSIONS Our results confirm that circulating AT1-AA are present significantly more often in preeclampsia than in normotensive pregnancy, however without association to acute atherosis. Whether circulating maternal AT1-AA or acute atherosis target young women at increased long-term cardiovascular risk warrants further investigations.
Collapse
|
177
|
Oxidative Stress in Preeclampsia and Placental Diseases. Int J Mol Sci 2018; 19:ijms19051496. [PMID: 29772777 PMCID: PMC5983711 DOI: 10.3390/ijms19051496] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/09/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia is a persistent hypertensive gestational disease characterized by high blood pressure and proteinuria, which presents from the second trimester of pregnancy. At the cellular level, preeclampsia has largely been associated with the release of free radicals by the placenta. Placenta-borne oxidative and nitrosative stresses are even sometimes considered as the major molecular determinants of the maternal disease. In this review, we present the recent literature evaluating free radical production in both normal and pathological placentas (including preeclampsia and other major pregnancy diseases), in humans and animal models. We then assess the putative effects of these free radicals on the placenta and maternal endothelium. This analysis was conducted with regard to recent papers and possible therapeutic avenues.
Collapse
|
178
|
Konečná B, Lauková L, Vlková B. Immune activation by nucleic acids: A role in pregnancy complications. Scand J Immunol 2018; 87:e12651. [PMID: 29479732 DOI: 10.1111/sji.12651] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 02/15/2018] [Indexed: 12/25/2022]
Abstract
Cell-free self-DNA or RNA may induce an immune response by activating specific sensing receptors. During pregnancy, placental nucleic acids present in the maternal circulation further activate these receptors due to the presence of unmethylated CpG islands. A higher concentration of cell-free foetal DNA is associated with pregnancy complications and a higher risk for foetal rejection. Cell-free foetal DNA originates from placental trophoblasts. It appears in different forms: free, bound to histones in nucleosomes, in neutrophil extracellular traps (NETs) and in extracellular vesicles (EVs). In several pregnancy complications, cell-free foetal DNA triggers the production of proinflammatory cytokines, and this production results in a cellular and humoral immune response. This review discusses preeclampsia, systemic lupus erythematosus, foetal growth restriction, gestational diabetes, rheumatoid arthritis and obesity in pregnancy from an immunological point of view and closely examines the different pathways that result in maternal inflammation. Understanding the role of cell-free nucleic acids, as well as the biogenesis of NETs and EVs, will help us to specify their functions or targets, which seem to be important in pregnancy complications. It is still not clear whether higher concentrations of cell-free nucleic acids in the maternal circulation are the cause or consequence of various complications. Therefore, further clinical studies and, even more importantly, animal experiments that focus on the involved immunological pathways are needed.
Collapse
Affiliation(s)
- B Konečná
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - L Lauková
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - B Vlková
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
179
|
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: 5.1] [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
|
180
|
Cunningham MW, Castillo J, Ibrahim T, Cornelius DC, Campbell N, Amaral L, Vaka VR, Usry N, Williams JM, LaMarca B. AT1-AA (Angiotensin II Type 1 Receptor Agonistic Autoantibody) Blockade Prevents Preeclamptic Symptoms in Placental Ischemic Rats. Hypertension 2018; 71:886-893. [PMID: 29555668 DOI: 10.1161/hypertensionaha.117.10681] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/16/2017] [Accepted: 02/08/2018] [Indexed: 01/02/2023]
Abstract
Women with preeclampsia produce AT1-AA (agonistic autoantibodies to the angiotensin II type 1 receptor), which stimulate reactive oxygen species, inflammatory factors, and hypertensive mechanisms (ET [endothelin] and sFlt-1 [soluble fms-like tyrosine kinase-1]) in rodent models of preeclampsia. The placental ischemic reduced uterine perfusion pressure (RUPP) rat model of preeclampsia exhibits many of these features. In this study, we examined the maternal outcomes of AT1-AA inhibition ('n7AAc') in RUPP rats. Blood pressure was higher in RUPP rats versus normal pregnant (NP) rats (123±2 versus 99±2 mm Hg, P<0.05), which was reduced in RUPP+'n7AAc' (105±3 versus 123±2 mm Hg, P<0.05 versus RUPP). Uterine artery resistant index was increased in RUPP versus NP rats (0.71±0.02 versus 0.49±0.02, P<0.05) and normalized in RUPP+'n7AAc' rats (0.55±0.03). Antiangiogenic factor sFlt-1 was elevated in RUPP versus NP rats (176±37 versus 77±15 pg/mL, P<0.05) but normalized in RUPP+'n7AAc' (86±9, P=0.05 versus RUPP). Plasma nitrate and nitrite were decreased (14±1 versus 20±1 µMNO3, P<0.05) and isoprostanes were elevated (20 117±6304 versus 2809±1375 pg/mL, P<0.05) in RUPP versus NP rats; and normalized in RUPP+'n7AAc' rats; (18±2 µMNO3; 4311±1 pg/mL). PPET-1 (preproendothelin-1) expression increased 4-fold in RUPP versus NP rats which were prevented with 'n7AAc'. Importantly, placental cytolytic natural killer cells were elevated in RUPP versus NP rats (8±2% versus 2±2% gated, P<0.05), which was prevented in RUPP+'n7AAc' total (3±1% gated, P<0.05) In conclusion, AT1-AA inhibition prevents the rise in maternal blood pressure and several pathophysiological factors associated with preeclampsia in RUPP rats and could be a potential therapy for preeclampsia.
Collapse
Affiliation(s)
- Mark W Cunningham
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Javier Castillo
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Tarek Ibrahim
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Denise C Cornelius
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Nathan Campbell
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Lorena Amaral
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Venkata Ramana Vaka
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Nathan Usry
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Jan M Williams
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson
| | - Babbette LaMarca
- From the Departments of Pharmacology and Toxicology (M.W.C., T.I., D.C.C., N.C., L.A., V.R.V., N.U., J.M.W., B.L.), Obstetrics and Gynecology (J.C., B.L.), and Emergency Medicine (D.C.C.), University of Mississippi Medical Center, Jackson.
| |
Collapse
|
181
|
Abstract
Preeclampsia is characterized by blood pressure greater than 140/90 mmHg in the second half of pregnancy. This disease is a major contributor to preterm and low birth weight babies. The early delivery of the baby, which becomes necessary for maintaining maternal well-being, makes preeclampsia the leading cause for preterm labor and infant mortality and morbidity. Currently, there is no cure for this pregnancy disorder. The current clinical management of PE is hydralazine with labetalol and magnesium sulfate to slow disease progression and prevent maternal seizure, and hopefully prolong the pregnancy. This review will highlight factors implicated in the pathophysiology of preeclampsia and current treatments for the management of this disease.
Collapse
|
182
|
Jünemann A, Hohberger B, Rech J, Sheriff A, Fu Q, Schlötzer-Schrehardt U, Voll RE, Bartel S, Kalbacher H, Hoebeke J, Rejdak R, Horn F, Wallukat G, Kunze R, Herrmann M. Agonistic Autoantibodies to the β2-Adrenergic Receptor Involved in the Pathogenesis of Open-Angle Glaucoma. Front Immunol 2018; 9:145. [PMID: 29483909 PMCID: PMC5816038 DOI: 10.3389/fimmu.2018.00145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/17/2018] [Indexed: 11/21/2022] Open
Abstract
Glaucoma is a frequent ocular disease that may lead to blindness. Primary open-angle glaucoma (POAG) and ocular hypertension (OHT) are common diseases with increased intraocular pressure (IOP), which are mainly responsible for these disorders. Their pathogenesis is widely unknown. We screened the sera of patients with POAG and OHT for the prevalence of autoantibodies (AAb) against G protein-coupled receptors (GPCRs) in comparison to controls. Employing frequency modulation of spontaneously contracting neonatal rat cardiomyocytes in vitro, agonistic GPCR AAb were to be detected in roughly 75% of the patients with POAG and OHT, however, not in controls. Using inhibitory peptides the AAb’ target was identified as β2 adrenergic receptor (β2AR). The AAb interact with the second extracellular loop of β2AR. The peptides 181–187 and 186–192 were identified as binding sites of the AAb within the extracellular loop II. The binding of the AAb to β2ARs was verified by surface-plasmon-resonance analysis. The isotype of the AAb was (immunoglobulin) IgG3. In an additional pilot principal-of-proof study, including four patients with POAG, the removal of the AAb against the β2AR and other immunoglobulins G by immunoadsorption resulted in a transient reduction of IOP. These findings might indicate a possible role of agonistic AAb directed against β2ARs in the dynamics of aqueous humor and might support a contribution of adaptive autoimmunity in the etiopathogenesis of POAG and OHT.
Collapse
Affiliation(s)
- Anselm Jünemann
- Department of Ophthalmology, University of Rostock, Rostock, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ahmed Sheriff
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Qin Fu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Reinhard Edmund Voll
- IZKF Research Group 2, Nikolaus-Fiebiger-Center of Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Bartel
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Hubert Kalbacher
- IFIB - Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Johan Hoebeke
- C.N.R.S. UPR 9021 «Chimie et Immunologie Thérapeutiques», Strasbourg, France
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Folkert Horn
- Department of Ophthalmology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gerd Wallukat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Kunze
- Science Office, Berlin-Buch, Campus Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Herrmann
- Department of Internal Medicine III, Institute of Clinical Immunology and Rheumatology, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
183
|
Taylor EB, Barati MT, Powell DW, Turbeville HR, Ryan MJ. Plasma Cell Depletion Attenuates Hypertension in an Experimental Model of Autoimmune Disease. Hypertension 2018; 71:719-728. [PMID: 29378858 DOI: 10.1161/hypertensionaha.117.10473] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/26/2017] [Accepted: 01/06/2018] [Indexed: 12/24/2022]
Abstract
Numerous studies show a direct relation between circulating autoantibodies, characteristic of systemic autoimmune disorders, and primary hypertension in humans. Whether these autoantibodies mechanistically contribute to the development of hypertension remains unclear. Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder characterized by aberrant immunoglobulin production, notably pathogenic autoantibodies, and is associated with prevalent hypertension, renal injury, and cardiovascular disease. Because plasma cells produce the majority of serum immunoglobulins and are the primary source of autoantibodies in SLE, we hypothesized that plasma cell depletion using the proteasome inhibitor bortezomib would lower autoantibody production and attenuate hypertension. Thirty-week-old female SLE (NZBWF1) and control (NZW [New Zealand White]) mice were injected IV with vehicle (0.9% saline) or bortezomib (0.75 mg/kg) twice weekly for 4 weeks. Bortezomib treatment significantly lowered the percentage of bone marrow plasma cells in SLE mice. Total plasma IgG and anti-dsDNA IgG levels were higher in SLE mice compared with control mice but were lowered by bortezomib treatment. Mean arterial pressure (mm Hg) measured in conscious mice by carotid artery catheter was higher in SLE mice than in control mice, but mean arterial pressure was significantly lower in bortezomib-treated SLE mice. Bortezomib also attenuated renal injury, as assessed by albuminuria and glomerulosclerosis, and reduced glomerular immunoglobulin deposition and B and T lymphocytes infiltration into the kidneys. Taken together, these data show that the production of autoantibodies by plasma cells mechanistically contributes to autoimmune-associated hypertension and suggests a potential role for patients with primary hypertension who have increased circulating immunoglobulins.
Collapse
Affiliation(s)
- Erin B Taylor
- From the Department of Physiology and Biophysics (E.B.T., M.J.R.) and Department of Pharmacology & Toxicology (H.R.T.), University of Mississippi Medical Center, Jackson; Department of Medicine, University of Louisville School of Medicine, KY (M.T.B., D.W.P.); and G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.J.R.)
| | - Michelle T Barati
- From the Department of Physiology and Biophysics (E.B.T., M.J.R.) and Department of Pharmacology & Toxicology (H.R.T.), University of Mississippi Medical Center, Jackson; Department of Medicine, University of Louisville School of Medicine, KY (M.T.B., D.W.P.); and G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.J.R.)
| | - David W Powell
- From the Department of Physiology and Biophysics (E.B.T., M.J.R.) and Department of Pharmacology & Toxicology (H.R.T.), University of Mississippi Medical Center, Jackson; Department of Medicine, University of Louisville School of Medicine, KY (M.T.B., D.W.P.); and G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.J.R.)
| | - Hannah R Turbeville
- From the Department of Physiology and Biophysics (E.B.T., M.J.R.) and Department of Pharmacology & Toxicology (H.R.T.), University of Mississippi Medical Center, Jackson; Department of Medicine, University of Louisville School of Medicine, KY (M.T.B., D.W.P.); and G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.J.R.)
| | - Michael J Ryan
- From the Department of Physiology and Biophysics (E.B.T., M.J.R.) and Department of Pharmacology & Toxicology (H.R.T.), University of Mississippi Medical Center, Jackson; Department of Medicine, University of Louisville School of Medicine, KY (M.T.B., D.W.P.); and G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.J.R.).
| |
Collapse
|
184
|
Pre-Eclampsia and Eclampsia: An Update on the Pharmacological Treatment Applied in Portugal. J Cardiovasc Dev Dis 2018; 5:jcdd5010003. [PMID: 29367581 PMCID: PMC5872351 DOI: 10.3390/jcdd5010003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/13/2022] Open
Abstract
Pre-eclampsia and eclampsia are two hypertensive disorders of pregnancy, considered major causes of maternal and perinatal death worldwide. Pre-eclampsia is a multisystemic disease characterized by the development of hypertension after 20 weeks of gestation, with the presence of proteinuria or, in its absence, of signs or symptoms indicative of target organ injury. Eclampsia represents the consequence of brain injuries caused by pre-eclampsia. The correct diagnosis and classification of the disease are essential, since the therapies for the mild and severe forms of pre-eclampsia are different. Thus, this review aims to describe the most advisable antepartum pharmacotherapy for pre-eclampsia and eclampsia applied in Portugal and based on several national and international available guidelines. Slow-release nifedipine is the most recommended drug for mild pre-eclampsia, and labetalol is the drug of choice for the severe form of the disease. Magnesium sulfate is used to prevent seizures caused by eclampsia. Corticosteroids are used for fetal lung maturation. Overall, the pharmacological prevention of these diseases is limited to low-dose aspirin, so it is important to establish the safest and most effective available treatment.
Collapse
|
185
|
Autoantibodies against angiotensin and adrenergic receptors: more than a biomarker? Clin Sci (Lond) 2018; 132:127-130. [PMID: 29326280 DOI: 10.1042/cs20171485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
Agonistic autoantibodies (AAs) directed against receptors of the sympathetic nervous system and the renin-angiotensin system have been suggested to contribute to cardiovascular and renal disease, in particular hypertension, preeclampsia, and graft failure in kidney transplantation patients. Consequently, they are now also being studied as biomarker for these conditions. This commentary summarizes our current understanding of these AAs, critically discussing whether they truly act as agonist, and focusing on the wide array of assays that are currently used for their quantification.
Collapse
|
186
|
Zhang X, Mirocha J, Aintablian T, Dimbil S, Moriguchi J, Arabia F, Kobashigawa JA, Reinsmoen N. Revealing a new mode of sensitization induced by mechanical circulatory support devices: Impact of anti-AT 1 R antibodies. Clin Transplant 2018; 32. [PMID: 29245181 DOI: 10.1111/ctr.13178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Increased levels of angiotensin II type 1 receptor (AT1 R) antibody have been shown to be associated with allograft rejection. This study aims to determine the rate of development of antibody to AT1 R after mechanical circulatory support device (MCS) implantation, and if the development of strong binding AT1 R antibodies is associated with survival. METHODS Eighty-eight patients who had one MCS implantation were accessed based on serum availability. Mechanical circulatory support devices in this cohort included pneumatic bilateral paracorporeal ventricular assist device, continuous flow left ventricular assist device, and total artificial heart. RESULTS Of 88 patients, seven patients had AT1 R antibodies ≥40 U/mL preimplantation. For 81 patients who had AT1 R antibodies <40 U/mL, the median value was 8 U/mL. Of these 81 patients, AT1 R antibody levels in 55 (68%) patients reached the saturated concentration (≥40 U/mL) postimplantation (P < .0001), with the highest percentage of patients with the saturated level of AT1 R antibody observed in the pneumatic bilateral paracorporeal ventricular assist device group. Compared to patients without the saturated level of AT1 R antibodies, patients with the saturated AT1 R antibody level had lower 18-month survival (P = .040). CONCLUSION Mechanical circulatory support devices implantation significantly increases AT1 R antibody levels. The saturated level of AT1 R antibodies is associated with lower patient survival postimplantation.
Collapse
Affiliation(s)
- Xiaohai Zhang
- HLA and Immunogenetics Laboratory, Comprehensive Transplant Center, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - James Mirocha
- Biostatistics Core, Cedars-Sinai Health System, Los Angeles, CA, USA
| | | | | | - Jaime Moriguchi
- Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Francisco Arabia
- Cardiovascular Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Nancy Reinsmoen
- HLA and Immunogenetics Laboratory, Comprehensive Transplant Center, Cedars-Sinai Health System, Los Angeles, CA, USA
| |
Collapse
|
187
|
|
188
|
Zeng L, Yang K, Ge J. Uncovering the Pharmacological Mechanism of Astragalus Salvia Compound on Pregnancy-Induced Hypertension Syndrome by a Network Pharmacology Approach. Sci Rep 2017; 7:16849. [PMID: 29203792 PMCID: PMC5715142 DOI: 10.1038/s41598-017-17139-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/19/2017] [Indexed: 01/08/2023] Open
Abstract
To uncover the pharmacological mechanism of Astragalus Salvia compound (ASC) on pregnancy-induced hypertension syndrome (PIH), to provide useful information for clinical, as well as to connect the basic and clinical by a network pharmacological approach, we used network pharmacological approach. We collected ASC's compounds by traditional Chinese Medicine databases, and input them into PharmMapper to got their targets. Then we acquired PIH targets from Genecards and OMIM, collected the interactions of all the targets and other human proteins via String and INACT. We also constructed the network by Cytoscape and analyze it by MCODE so as to get clusters. Finally, we put all the targets of clusters into DAVID to do GO enrichment analysis. After these, four networks are constructed by Cytoscape; they are PIH network, compound-compound target network of ASC, ASC-PIH network, and compound target-PIH target-other human proteins' PPI network. According to the results, we think that ASC may directly regulate several biological processes and their genes in "endothelial cell activation and injury" and "placental or trophoblast cell ischemia" models to treat PIH. And it may indirectly act on the rest of the biological process to treat PIH or may not.
Collapse
Affiliation(s)
- Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Kailin Yang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Jinwen Ge
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China.
| |
Collapse
|
189
|
Long-term presence of angiotensin II type 1 receptor autoantibody reduces aldosterone production by triggering Ca2+ overload in H295R cells. Immunol Res 2017; 66:44-51. [DOI: 10.1007/s12026-017-8963-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
190
|
Szpera-Gozdziewicz A, Gozdziewicz T, Wirstlein P, Wender-Ozegowska E, Breborowicz GH. The agonistic autoantibodies to the angiotensin II type 1 receptor in pregnancies complicated by hypertensive disorders. J Matern Fetal Neonatal Med 2017; 32:1219-1223. [PMID: 29092665 DOI: 10.1080/14767058.2017.1400006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: The etiology and pathogenesis of pregnancy-related hypertensive disorders is complex and multifactorial. The aim of our study is the investigation of the differences in the autoantibodies against angiotensin II type 1 receptor (AT1-AA) titers among pregnant patients with chronic hypertension, gestational hypertension, and preeclampsia compared to the healthy pregnant women. Patients and methods: We created three study groups (preeclampsia [n = 16], chronic hypertension [n = 13], gestational hypertension [n = 17]) and the control group consisting of 17 healthy pregnant women. Every compared group was matched for mother's age, parity, prepregnancy BMI, and gestational age at time of recruitment into study. The autoantibodies titer were assessed using commercially available ELISA kit. Results: We found a statistically higher AT1-AA titer in the group of patients with gestational hypertension (GH) and preeclampsia (PE) compared to healthy normotensive pregnant women (median 9.6 versus 7.8 ng/ml, p = .01 and 10.9 ng/ml versus 7.8 ng/ml, p = .02, respectively). There was no correlation between blood pressure values and AT1-AA titer in any group. We found no correlation in group with preeclampsia between urinary protein excretion and AT1-AA titer (p = .23, R = 0.32). Conclusions: We assume that pregnancy-related hypertensive disorders might be autoimmune diseases and AT1-AA contribute to the pathophysiology of the disease. Our study may have some therapeutic implications and shows the necessity of new research into the mechanisms involved in the production of AT1-AA. Such investigations might enable to inhibit the formation of these autoantibodies or elaborate another method for AT1-AA removal.
Collapse
Affiliation(s)
- Agata Szpera-Gozdziewicz
- a Department of Perinatology and Gynecology, Division of Perinatology and Gynecology , Poznan University of Medical Sciences , Poznań , Poland
| | - Tomasz Gozdziewicz
- b Department of Perinatology and Gynecology, Division of Gynecology , Poznan University of Medical Sciences , Poznań , Poland
| | - Przemysław Wirstlein
- c Department of Obstetrics, Division of Reproduction, Gynecology and Gynecologic Oncology , Poznan University of Medical Sciences , Poznań , Poland
| | - Ewa Wender-Ozegowska
- c Department of Obstetrics, Division of Reproduction, Gynecology and Gynecologic Oncology , Poznan University of Medical Sciences , Poznań , Poland
| | - Grzegorz H Breborowicz
- a Department of Perinatology and Gynecology, Division of Perinatology and Gynecology , Poznan University of Medical Sciences , Poznań , Poland
| |
Collapse
|
191
|
Moroncini G, Svegliati Baroni S, Gabrielli A. Agonistic antibodies in systemic sclerosis. Immunol Lett 2017; 195:83-87. [PMID: 29032187 DOI: 10.1016/j.imlet.2017.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/05/2017] [Accepted: 10/11/2017] [Indexed: 10/18/2022]
Abstract
Systemic sclerosis (SSc) is characterized by microangiopathy, excessive fibrosis, and the presence of circulating autoantibodies to several cellular and extracellular components. The role of autoimmunity in generating the clinical and pathologic phenotypes in SSc has been long debated and is still matter of controversy. Distinct specificities of antinuclear antibodies (ANAs) are selectively detected in SSc patients and are associated with unique disease manifestations, but do not have a proven pathogenic role. A new group of autoantibodies reactive with cell surface receptors have been identified in SSc patients. They have been shown to directly activate pathways that may contribute to tissue and vascular damage. As such, they are proposed to have a role as agonistic autoantibodies in SSc. According to Koch's third postulate, the autoantibodies in question should cause disease when introduced into a healthy subject. Therefore, our review will focus on those autoantibodies for which agonistic activity has already been demonstrated not only in vitro, but, at least partly, also in vivo. These include the antibodies anti-endothelial cells (AECA), anti-Platelet-Derived Growth Factor Receptor (PDGFR), anti-Angiotensin II type 1 receptor (AT1R) and anti-endothelin-1 type A receptor (ETaR). In this review, we will discuss also a class of antagonistic autoantibodies, the anti-muscarinic-3 receptor (M3R) antibodies, since they seem to fulfill the aforementioned requirements.
Collapse
Affiliation(s)
- Gianluca Moroncini
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, 60126, Italy
| | - Silvia Svegliati Baroni
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, 60126, Italy
| | - Armando Gabrielli
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, 60126, Italy.
| |
Collapse
|
192
|
Jafri S, Ormiston ML. Immune regulation of systemic hypertension, pulmonary arterial hypertension, and preeclampsia: shared disease mechanisms and translational opportunities. Am J Physiol Regul Integr Comp Physiol 2017; 313:R693-R705. [PMID: 28978513 DOI: 10.1152/ajpregu.00259.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022]
Abstract
Systemic hypertension, preeclampsia, and pulmonary arterial hypertension (PAH) are diseases of high blood pressure in the systemic or pulmonary circulation. Beyond the well-defined contribution of more traditional pathophysiological mechanisms, such as changes in the renin-angiotensin-aldosterone system, to the development of these hypertensive disorders, there is substantial clinical evidence supporting an important role for inflammation and immunity in the pathogenesis of each of these three conditions. Over the last decade, work in small animal models, bearing targeted deficiencies in specific cytokines or immune cell subsets, has begun to clarify the immune-mediated mechanisms that drive changes in vascular structure and tone in hypertensive disease. By summarizing the clinical and experimental evidence supporting a contribution of the immune system to systemic hypertension, preeclampsia, and PAH, the current review highlights the cellular and molecular pathways that are common to all three hypertensive disorders. These mechanisms are centered on an imbalance in CD4+ helper T cell populations, defined by excessive Th17 responses and impaired Treg activity, as well as the excessive activation or impairment of additional immune cell types, including macrophages, dendritic cells, CD8+ T cells, B cells, and natural killer cells. The identification of common immune mechanisms in systemic hypertension, preeclampsia, and PAH raises the possibility of new therapeutic strategies that target the immune component of hypertension across multiple disorders.
Collapse
Affiliation(s)
- Salema Jafri
- University of Cambridge, Department of Medicine, Cambridge, United Kingdom; and
| | - Mark L Ormiston
- Queen's University, Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Kingston, Canada
| |
Collapse
|
193
|
Coppo P. [Secondary thrombotic microangiopathies]. Rev Med Interne 2017; 38:731-736. [PMID: 28890263 DOI: 10.1016/j.revmed.2017.06.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 10/18/2022]
Abstract
Thrombotic microangiopathies (TMA) are termed secondary when associated to a specific context favouring their occurrence. They encompass mainly TMA associated with pregnancy, allogeneic hematopoietic stem cell transplantation, cancer, drugs, or HIV infection. Secondary TMA represent a heterogeneous group of diseases which clinical presentation largely depends on the associated context. It is therefore mandatory to recognize these conditions since they have a significant impact in TMA management and prognosis. A successful management still represents a challenge in secondary TMA. Significant progresses have been made in the understanding of pregnancy-associated TMA, allowing an improvement of prognosis; on the opposite, other forms of secondary TMA such as hematopoietic stem cell transplantation-associated TMA or TMA associated with chemotherapy remain of dismal prognosis. A better understanding of pathophysiology in these forms of TMA, in association with a more empirical approach through the use of new therapeutic agents that can also help in the understanding on new mechanisms a posteriori, should improve their prognosis. The preliminary encouraging results reported with complement blockers in this field could represent a convincing example.
Collapse
Affiliation(s)
- P Coppo
- Service d'hématologie, centre de référence des microangiopathies thrombotiques, hôpital Saint-Antoine, UPMC université Paris 6, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France.
| | -
- Service d'hématologie, centre de référence des microangiopathies thrombotiques, hôpital Saint-Antoine, UPMC université Paris 6, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France
| |
Collapse
|
194
|
Armanini D, Andrisani A, Donà G, Bordin L, Ambrosini G, Sabbadin C. Hypothesis on a relationship between hyperaldosteronism, inflammation, somatic mutations, and autoimmunity. J Clin Hypertens (Greenwich) 2017; 19:1060-1062. [DOI: 10.1111/jch.13074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Decio Armanini
- Department of Medicine, Endocrinology (DIMED); University of Padua; Padua Italy
| | | | - Gabriella Donà
- Department of Medicine, Endocrinology (DIMED); University of Padua; Padua Italy
| | - Luciana Bordin
- Department of Molecular Medicine, Biological Chemistry; University of Padua; Padua Italy
| | - Guido Ambrosini
- Department of Women's Health, Salus Pueri; University of Padua; Padua Italy
| | - Chiara Sabbadin
- Department of Medicine, Endocrinology (DIMED); University of Padua; Padua Italy
| |
Collapse
|
195
|
Kaculini E, Idrizi L, Duli M, Koroshi A, Shehu A, Spahia N, Barbullushi M. Preeclampsia: from Pathophysiology to Treatment. BANTAO JOURNAL 2017. [DOI: 10.1515/bj-2016-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Preeclampsia is a multisystem disorder unique to human pregnancy and is its most common glomerular complication. It occurs in 2% to 8% of pregnancies and is a major contributor to maternal mortality worldwide. Although the pathophysiology of this syndrome is not fully understood, many pathogenetic mechanisms are involved in this disorder. The role of the placenta is crucial in the development of this disorder. Some pathogenetic mechanisms involved in this disease comprise defective deep placentation, autoantibodies to type-1 angiotensin II receptor, endothelial dysfunction, oxidative stress, platelet and thrombin activation, intravascular inflammation, and the imbalance between angiogenic and antiangiogenic factors which is thought to be one of the most crucial mechanisms. Further understanding of the full picture could enhance our current knowledge of the pathogenesis of preeclampsia and improve its treatment. Thus, based on specific biomarkers the diagnosis and subclassification of preeclampsia might be more accurate in identifying patients at risk, monitoring disease progression and providing effective interventions
Collapse
Affiliation(s)
- Enton Kaculini
- Faculty of Medical Sciences, Albanian University, Tirana , Albania
| | - lma Idrizi
- Department of Internal Medicine, Service of Nephrology, University Hospital Center “Mother Teresa”, Tirana , Albania
| | - Marsida Duli
- Faculty of Medical Sciences, Albanian University, Tirana , Albania
| | - Alketa Koroshi
- Department of Internal Medicine, Service of Nephrology, University Hospital Center “Mother Teresa”, Tirana , Albania
| | - Alma Shehu
- Medical Representative, Novartis Pharma, Tirana , Albania
| | - Nereida Spahia
- Department of Internal Medicine, Service of Nephrology, University Hospital Center “Mother Teresa”, Tirana , Albania
| | - Myftar Barbullushi
- Department of Internal Medicine, Service of Nephrology, University Hospital Center “Mother Teresa”, Tirana , Albania
| |
Collapse
|
196
|
Liu C, Kellems RE, Xia Y. Inflammation, Autoimmunity, and Hypertension: The Essential Role of Tissue Transglutaminase. Am J Hypertens 2017; 30:756-764. [PMID: 28338973 PMCID: PMC5861548 DOI: 10.1093/ajh/hpx027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022] Open
Abstract
Inflammatory cytokines cause hypertension when introduced into animals. Additional evidence indicates that cytokines induce the production of autoantibodies that activate the AT1 angiotensin receptor (AT1R). Extensive evidence shows that these autoantibodies, termed AT1-AA, contribute to hypertension. We review here recent studies showing that cytokine-induced hypertension and AT1-AA production require the ubiquitous enzyme, tissue transglutaminase (TG2). We consider 3 mechanisms by which TG2 may contribute to hypertension. (i) One involves the posttranslational modification (PTM) of AT1Rs at a glutamine residue that is present in the epitope sequence (AFHYESQ) recognized by AT1-AA. (ii) Another mechanism by which TG2 may contribute to hypertension is by PTM of AT1Rs at glutamine 315. Modification at this glutamine prevents ubiquitination-dependent proteasome degradation and allows AT1Rs to accumulate. Increased AT1R abundance is likely to account for increased sensitivity to Ang II activation and in this way contribute to hypertension. (iii) The increased TG2 produced as a result of elevated inflammatory cytokines is likely to contribute to vascular stiffness by modification of intracellular contractile proteins or by crosslinking vascular proteins in the extracellular matrix. This process, termed inward remodeling, results in reduced vascular lumen, vascular stiffness, and increased blood pressure. Based on the literature reviewed here, we hypothesize that TG2 is an essential participant in cytokine-induced hypertension. From this perspective, selective TG2 inhibitors have the potential to be pharmacologic weapons in the fight against hypertension.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| | - Rodney E. Kellems
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, McGovern Medical School of the University of Texas at Houston, Houston, Texas, USA
| |
Collapse
|
197
|
Moe K, Heidecke H, Dechend R, Staff AC. Dysregulation of circulating autoantibodies against VEGF-A, VEGFR-1 and PlGF in preeclampsia - A role in placental and vascular health? Pregnancy Hypertens 2017; 10:83-89. [PMID: 29153696 DOI: 10.1016/j.preghy.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/31/2017] [Accepted: 06/04/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Preeclampsia is a state of antiangiogenesis, with high levels of maternal circulating sVEGFR-1 (soluble vascular endothelial growth factor receptor 1, also named sFlt1) and low levels of PlGF (placenta growth factor). Various autoantibodies have been detected in preeclamptic patients. We hypothesize that circulating autoantibodies against VEGF-A (AA-VEGF-A), VEGFR-1 (AA-VEGFR-1) and PlGF (AA-PlGF) are present in preeclamptic women, with different levels from pregnant women with normotensive pregnancies. Secondly, we wanted to analyze if autoantibody levels are associated to sFlt1 or PLGF levels. STUDY DESIGN Retrospective cross sectional study of 88 women with singleton pregnancies who delivered at Oslo University Hospital of whom 46 had preeclampsia and 42 had uncomplicated normotensive pregnancies. Novel immunoassays for IgG-autoantibodies against VEGFA, VEGFR-1 and PlGF were developed and serum samples were assayed. MAIN OUTCOME MEASURES AND RESULTS AA-VEGF-A, AA-VEGF-R1 and AA-PlGF were significantly lower in preeclamptic pregnancies (n=42) compared to normotensive pregnancies (n=46) (p<0.05). On unadjusted analysis, only AA-VEGFA and AA-VEGFR-1 were predictors of PE, but none were independent predictors after adjusting for BMI (body mass index) and parity. In the subgroup of normotensive and PE women with overlapping sVEGFR-1/PlGF-ratios, AA-VEGF was a significant predictor of PE with AUC: 0.735. CONCLUSION IgG autoantibodies against VEGF-A VEGFR-1 and PlGF can be found in pregnant women. They are dysregulated in preeclampsia. The roles of these autoantibodies are unknown, but this study suggests they play a protective role in pregnancy. The levels of AA against VEGF-A, VEGFR-1 and PlGF might be important factors contributing to anti-angiogenesis regulation.
Collapse
Affiliation(s)
- Kjartan Moe
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway, PB 1171, Blindern, 0381 Oslo, Norway.
| | - Harald Heidecke
- CellTrend GmbH, Im Biotechnologiepark, 14943 Luckenwalde, Germany.
| | - Ralf Dechend
- HELIOS Clinic, Berlin, Germany; Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbrueck Center for Molecular Medicine, Berlin, Germany.
| | - Anne Cathrine Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway, PB 1171, Blindern, 0381 Oslo, Norway; Department of Obstetrics and Gynaecology, Oslo University Hospital, PB 4956 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
198
|
Stanhewicz AE, Jandu S, Santhanam L, Alexander LM. Increased Angiotensin II Sensitivity Contributes to Microvascular Dysfunction in Women Who Have Had Preeclampsia. Hypertension 2017; 70:382-389. [PMID: 28652473 DOI: 10.1161/hypertensionaha.117.09386] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/03/2017] [Accepted: 05/01/2017] [Indexed: 01/13/2023]
Abstract
Women who have had preeclampsia have increased cardiovascular disease risk; however, the mechanism(s) responsible for this association remain unclear. Microvascular damage sustained during a preeclamptic pregnancy may persist postpartum. The putative mechanisms mediating this dysfunction include a reduction in NO-dependent dilation and an increased sensitivity to angiotensin II. In this study, we evaluated endothelium-dependent dilation, angiotensin II sensitivity, and the therapeutic effect of angiotensin II receptor blockade (losartan) on endothelium-dependent dilation in vivo in the microvasculature of women with a history of preeclampsia (n=12) and control women who had a healthy pregnancy (n=12). We hypothesized that preeclampsia would have (1) reduced endothelium-dependent dilation, (2) reduced NO-mediated dilation, and (3) increased sensitivity to angiotensin II. We further hypothesized that localized losartan would increase endothelium-dependent vasodilation in preeclampsia. We assessed microvascular endothelium-dependent vasodilator function by measurement of cutaneous vascular conductance responses to graded infusion of acetylcholine (acetylcholine; 10-7-102 mmol/L) and a standardized local heating protocol in control sites and sites treated with 15 mmol/L L-NAME (NG-nitro-l-arginine methyl ester; NO-synthase inhibitor) or 43 µmol/L losartan. Further, we assessed microvascular vasoconstrictor sensitivity to angiotensin II (10-20-10-4 mol/L). Preeclampsia had significantly reduced endothelium-dependent dilation (-0.3±0.5 versus -1.0±0.4 logEC50; P<0.001) and NO-dependent dilation (16±3% versus 39±6%; P=0.006). Preeclampsia also had augmented vasoconstrictor sensitivity to angiotensin II (-10.2±1.3 versus -8.3±0.5; P=0.006). Angiotensin II type I receptor inhibition augmented endothelium-dependent vasodilation and NO-dependent dilation in preeclampsia but had no effect in healthy pregnancy. These data suggest that women who have had preeclampsia have persistent microvascular dysfunction postpartum, mediated, in part, by increased sensitivity to angiotensin II.
Collapse
Affiliation(s)
- Anna E Stanhewicz
- From the Department of Kinesiology, Pennsylvania State University, University Park, (A.E.S., L.M.A.); and Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (S.J., L.S.).
| | - Sandeep Jandu
- From the Department of Kinesiology, Pennsylvania State University, University Park, (A.E.S., L.M.A.); and Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (S.J., L.S.)
| | - Lakshmi Santhanam
- From the Department of Kinesiology, Pennsylvania State University, University Park, (A.E.S., L.M.A.); and Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (S.J., L.S.)
| | - Lacy M Alexander
- From the Department of Kinesiology, Pennsylvania State University, University Park, (A.E.S., L.M.A.); and Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (S.J., L.S.)
| |
Collapse
|
199
|
|
200
|
Ueki N, Kanasaki K, Kanasaki M, Takeda S, Koya D. Catechol-O-Methyltransferase Deficiency Leads to Hypersensitivity of the Pressor Response Against Angiotensin II. Hypertension 2017; 69:1156-1164. [DOI: 10.1161/hypertensionaha.117.09247] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 11/16/2022]
Abstract
Catechol-O-methyltransferase (COMT) metabolizes 2-hydroxyestradiol into 2-methoxyestradiol (2-ME); COMT deficiency has shown to be associated with hypertension in men and preeclampsia, the disease associated with hypersensitivity of pressor response against angiotensin II (Ang II). Here, we found that COMT deficiency could explain the hypersensitivity of pressor response against Ang II in mice because of the lack of 2-ME–dependent suppression of angiotensin II receptor type 1 (AT1R). Male C57BL/6 mice were subjected to COMT inhibitor (COMTi: 25 mg/kg per day) or oil (control) for 4 weeks, with or without low-dose Ang II infusion (ANGII: 70 ng/kg per minute) for the last 3 weeks. The Ang II–infused mice were treated with 2-ME (10 ng/d) or vehicle for the last 1 week. We obtained the following experimental groups: control, ANGII, COMTi, COMTi+ANGII, and COMTi+ANGII+2-ME. We performed similar experiments using the in vivo administration of small interfering RNA of COMT instead of COMTi. Neither ANGII nor COMTi exhibited significant alterations in systolic blood pressure. Compared with ANGII or COMTi, COMTi+ANGII displayed significantly higher systolic blood pressure, albuminuria, and glomerular endotheliosis; 2-ME normalized such alterations. Similar phenotypes were observed in COMT small interfering RNA–treated mice. In the aorta of COMT-deficient mice, AT1R expression was increased; 2-ME suppressed AT1R expression. The 2-ME exhibited peroxisome proliferator–activated receptor γ agonistic activity in vitro and ex vivo plasma from pregnant female mice as well. In vitro, 2-ME suppressed both basal and Ang II–induced AT1R levels in a peroxisome proliferator–activated receptor γ–dependent manner. The 2-ME is relevant to combat COMT deficiency–associated hypertensive disorders via suppression of AT1R by its peroxisome proliferator–activated receptor γ activity.
Collapse
Affiliation(s)
- Norikazu Ueki
- From the Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan (N.U., S.T.); and Department of Diabetology and Endocrinology (N.U., K.K., M.K., D.K.) and Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (K.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Keizo Kanasaki
- From the Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan (N.U., S.T.); and Department of Diabetology and Endocrinology (N.U., K.K., M.K., D.K.) and Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (K.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Megumi Kanasaki
- From the Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan (N.U., S.T.); and Department of Diabetology and Endocrinology (N.U., K.K., M.K., D.K.) and Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (K.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Satoru Takeda
- From the Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan (N.U., S.T.); and Department of Diabetology and Endocrinology (N.U., K.K., M.K., D.K.) and Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (K.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| | - Daisuke Koya
- From the Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan (N.U., S.T.); and Department of Diabetology and Endocrinology (N.U., K.K., M.K., D.K.) and Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute (K.K., D.K.), Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|