1
|
Rodriguez-Perez AI, Serrano-Heras G, Labandeira CM, Camacho-Meño L, Castro-Robles B, Suarez-Quintanilla JA, Muñoz-López M, Piqueras-Landete P, Guerra MJ, Segura T, Labandeira-Garcia JL. Serum angiotensin type-1 receptor autoantibodies and neurofilament light chain as markers of neuroaxonal damage in post-COVID patients. Front Immunol 2025; 16:1571027. [PMID: 40330487 PMCID: PMC12052551 DOI: 10.3389/fimmu.2025.1571027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Dysregulation of autoimmune responses and the presence of autoantibodies (AA), particularly those related to the renin-angiotensin system (RAS), have been implicated in the acute phase of COVID-19, and persistent dysregulation of brain RAS by RAS-related autoantibodies may also contribute to neurological symptoms of post-COVID. Methods We analyzed levels of serum and CSF RAS AA in post-COVID patients with neurological symptoms, individuals who have fully recovered from COVID-19 (after-COVID controls), and uninfected individuals, and their possible correlations with the serum marker of neuroaxonal damage neurofilament light chain (NfL) and the degrees of cognitive deficit. Results Both in serum and CSF, levels of AA agonists of the pro-inflammatory angiotensin II type 1 receptors (AT1-AA) were significantly elevated in this cohort of neurological post-COVID patients compared to both uninfected and after-COVID controls and correlated with serum levels of NfL. Changes in serum and CSF levels of AA promoting the RAS anti-inflammatory axis (upregulation of AA agonists of AT2 and Mas receptors, downregulation of AA antagonists of ACE2) suggest upregulation of the RAS compensatory response in this cohort of neurological post-COVID patients. Post-COVID patients with more pronounced cognitive impairment exhibited significantly higher CSF levels of MasR-AA and a trend toward elevated AT2-AA. Persistent brain RAS dysregulation, particularly persistent increase in AT1-AA, and its correlation with neuroaxonal damage markers and cognitive impairment, may play a significant role in neurological symptoms associated with post-COVID. Serum levels of NfL and AT1-AA may be interesting biomarkers for the early identification of CNS involvement in patients with neurological symptoms and a history of COVID-19. However, post-COVID is a highly heterogeneous entity and may result from various underlying mechanisms. The present study includes a cohort, which may differ from other cohorts with different clinical profiles, which may show different results on NfLs and CSF RAS autoantibodies, particularly AT1-AA. Conclusion These findings highlight the potential of targeting AT1 receptors as a therapeutic strategy for mitigating cognitive deficits in post-COVID patients showing upregulated AT1-AA levels.
Collapse
Affiliation(s)
- Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Research Health Institute of Santiago (IDIS), Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | | | - Laura Camacho-Meño
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Juan A. Suarez-Quintanilla
- Research Health Institute of Santiago (IDIS), Santiago de Compostela, Spain
- Primary Health-Care Unit Fontiñas, IDIS, University of Santiago de Compostela, Santiago de Compostela, La Coruña, Spain
| | - Mónica Muñoz-López
- Faculty of Medicine, University of Castilla-La Mancha (UCLM), Albacete, Spain
- Biomedicine Institute (IB-UCLM), Albacete, Spain
| | | | - María J. Guerra
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Research Health Institute of Santiago (IDIS), Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Tomas Segura
- Faculty of Medicine, University of Castilla-La Mancha (UCLM), Albacete, Spain
- Biomedicine Institute (IB-UCLM), Albacete, Spain
- Department of Neurology, General University Hospital of Albacete, Albacete, Spain
| | - José L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Research Health Institute of Santiago (IDIS), Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
2
|
Ponthier L, El Hamel C, Coste Mazeau P, Martinez S, Wehbe S, Froget R, Yardin C, Guigonis V. Angiotensin II type-1 receptor autoantibodies and effects in neonates of women with preeclampsia. BMC Pregnancy Childbirth 2025; 25:1. [PMID: 39748304 PMCID: PMC11697507 DOI: 10.1186/s12884-024-07102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Maternal agonistic autoantibodies against the angiotensin II type 1 receptor (AT1-AAs) have been implicated in the pathophysiology of preeclampsia, but their presence in their offsprings and their possible neonatal effects have not been specifically explored. This prospective study aimed to evaluate the presence of AT1-AAs and their potential clinical effects in neonates of AT1-AAs positive mothers. METHODS Women with preeclampsia and their neonates were included. Blood samples were collected in order to search for AT1-AAs. RESULTS AT1-AA determination was positive in 35 out of 64 of the studied women (54.7%). Thirty one newborns from the group of AT1-AA positive women were included and 22 (71%) were AT1-AA positive. The mothers' and children's AT1-AAs titers were significantly correlated. The 33 newborns from the group of AT1-AA negative women were all negative for AT1-AAs. Regarding the clinical data of newborns (birth weight, percentile of weight, gestational age, Apgar score at five minutes, mechanical or noninvasive ventilation), no significant difference was observed between the children with or without detected AT1-AAs. CONCLUSION Even though AT1-AAs are detected in newborns of positive mothers during the first days of life, no specific clinical signs seem to be associated with the presence of these antibodies.
Collapse
Affiliation(s)
- Laure Ponthier
- Collection Biologique de L'Hôpital de La Mère Et de L'Enfant CB-HME (Mother and Child Biobank), University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France.
- Department of Pediatrics, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France.
| | - Chahrazed El Hamel
- Collection Biologique de L'Hôpital de La Mère Et de L'Enfant CB-HME (Mother and Child Biobank), University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
- Department of Pediatrics, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| | - Perrine Coste Mazeau
- Department of Obstetric Gynecology, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| | - Sophie Martinez
- Department of Obstetric Gynecology, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| | - Sarah Wehbe
- Department of Obstetric Gynecology, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| | - Rachel Froget
- Department of Pediatrics, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
- INSERM CIC 1435, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| | - Catherine Yardin
- Collection Biologique de L'Hôpital de La Mère Et de L'Enfant CB-HME (Mother and Child Biobank), University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
- Department of Cytogenetics and Clinical Genetics, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
- UMR 7252, Limoges University, CNRS, XLIM, Limoges, France
| | - Vincent Guigonis
- Collection Biologique de L'Hôpital de La Mère Et de L'Enfant CB-HME (Mother and Child Biobank), University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
- Department of Pediatrics, University Hospital Center, 8 Avenue Dominique Larrey, Limoges, France
| |
Collapse
|
3
|
van Oostveen WM, Hoekstra EM, Levarht EWN, Kotliar IB, Sakmar TP, Toes REM, de Vries-Bouwstra JK, Heitman LH, Fehres CM. Absence of Functional Autoantibodies Targeting Angiotensin II Receptor Type 1 and Endothelin-1 Type A Receptor in Circulation and Purified IgG From Patients With Systemic Sclerosis. Arthritis Rheumatol 2024. [PMID: 39721751 DOI: 10.1002/art.43099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a rare but severe autoimmune disease characterized by immune dysregulation, fibrosis, and vasculopathy. Although previous studies have highlighted the presence of functional autoantibodies targeting the angiotensin II receptor type 1 (AT1) and endothelin-1 type A receptor (ETAR), leading to autoantibody-mediated receptor stimulation and subsequent activation of endothelial cells (ECs), a comprehensive understanding of the direct interaction between these autoantibodies and their receptors is currently lacking. Moreover, existing data confirming the presence of these autoantibodies in SSc often rely on similar methodologies and assays. Our aim was to replicate previous findings and to investigate the functional effects of IgG derived from patients with SSc (SSc IgG) on AT1 and ETAR signaling, the downstream EC response, and the presence of AT1-binding autoantibodies in circulation. METHODS Quantitative polymerase chain reaction and cytokine enzyme-linked immunosorbent assay, alongside a real-time cell analyzer, were used to assess receptor-specific functional characteristics of purified SSc IgG (n = 18). Additionally, a novel protein capture assay using solubilized epitope-tagged AT1 was developed to detect AT1-binding autoantibodies in plasma samples from patients with SSc (n = 28) and healthy donors (n = 14). RESULTS No evidence for EC activation in an AT1- or ETAR-dependent manner was revealed. Furthermore, stimulation with SSc IgG did not induce receptor activation or alter G protein-coupled receptor signaling on agonist stimulation in a model with receptor overexpression. Lastly, no AT1-binding autoantibodies were detected in plasma samples from patients with SSc when using epitope-tagged solubilized AT1. CONCLUSION Overall, our study did not provide evidence to support the presence of AT1- or ETAR-activating autoantibodies in purified SSc IgG or AT1-binding autoantibodies in the circulation of patients with SSc.
Collapse
Affiliation(s)
| | - Eva M Hoekstra
- Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ilana B Kotliar
- The Rockefeller University and Tri-Institutional PhD Program in Chemical Biology, New York, New York
| | | | - René E M Toes
- Leiden University Medical Center, Leiden, The Netherlands
| | | | - Laura H Heitman
- Leiden University and Oncode Institute, Leiden, The Netherlands
| | | |
Collapse
|
4
|
Barbosa GSB, Câmara NOS, Ledesma FL, Duarte Neto AN, Dias CB. Vascular injury in glomerulopathies: the role of the endothelium. FRONTIERS IN NEPHROLOGY 2024; 4:1396588. [PMID: 39780910 PMCID: PMC11707422 DOI: 10.3389/fneph.2024.1396588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In glomerulopathies, endothelial dysfunction and the presence of histological vascular lesions such as thrombotic microangiopathy, arteriolar hyalinosis, and arteriosclerosis are related to a severe clinical course and worse renal prognosis. The endothelial cell, which naturally has anti-inflammatory and anti-thrombotic regulatory mechanisms, is particularly susceptible to damage caused by various etiologies and can become dysfunctional due to direct/indirect injury or a deficiency of protective factors. In addition, endothelial regulation and protection involve participation of the complement system, factors related to angiogenesis, the renin-angiotensin system (RAS), endothelin, the glycocalyx, the coagulation cascade, interaction between these pathways, interactions between glomerular structures (the endothelium, mesangium, podocyte, and basement membrane) and interstitial structures (tubules, arterioles and small vessels). Dysregulation of those components is also associated with the progression of renal fibrosis, since endothelial cell damage promotes endothelial-to-mesenchymal transition. Although the potential mechanisms of vascular injury have been widely described in diabetic kidney disease, hypertensive nephrosclerosis, and hemolytic uremic syndrome, they require further elucidation in other glomerulopathies. A better understanding of the pathogenesis of vascular injury in patients with glomerular diseases could contribute to the development of specific treatments for such injury.
Collapse
Affiliation(s)
- Géssica Sabrine Braga Barbosa
- Renal Pathophysiology Laboratory, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | | | | | - Cristiane Bitencourt Dias
- Renal Pathophysiology Laboratory, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
5
|
Li X, Yang G, Ren J, Li X, Chen Y, Zhang Y, Shi Y, Yang X. Angiotensin II type-1 receptor autoantibody positively correlates with the rate of metaphase I oocytes in infertility with ovulatory disorder. J Reprod Immunol 2024; 166:104327. [PMID: 39255543 DOI: 10.1016/j.jri.2024.104327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/12/2024]
Abstract
The renin-angiotensin system (RAS) plays an important role in reproductive function. Our previous study identified that angiotensin II type-1 receptor autoantibody (AT1-AA), an autoantibody that activates RAS, was closely associated with infertility. However, its distribution in different types of infertility remained unclear. This study was designed to explore the distribution of AT1-AA in infertile patients and the connections between AT1-AA and oocyte development and pregnancy outcome. A total of 184 infertile women participated, with samples collected from peripheral venous blood. ELISA was used to detect AT1-AA levels in their sera. It was observed that the proportion of ovulation-disorder factors in AT1-AA-positive group was significantly higher than that in negative group (P=0.001). In 59 infertile women with ovulatory disorders, compared with negative group, AT1-AA-positive group had lower rate of retrieval (P=0.032) and metaphase II (MII) oocytes (P=0.011) but higher proportion of metaphase I (MI) oocytes (P=0.019). A negative correlation was found between the levels of AT1-AA and rate of retrieval and MII oocytes (P=0.027; P=0.043), whereas a positive correlation was observed with the proportion of MI oocytes (P=0.002). Moreover, a specific predictive value for proportion of reaching MII and MI oocytes was exhibited by AT1-AA (P < 0.01; P < 0.05). But no significant difference in embryonic parameters or pregnancy outcomes between two groups was observed (P > 0.05). This study revealed that serum AT1-AA levels were significantly increased in infertile women with ovulatory disorders and positively correlated with proportion of MI oocytes, but not associated with outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Xuemin Li
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China
| | - Guifang Yang
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China
| | - Jie Ren
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China
| | - Xiaonuo Li
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China
| | - Yao Chen
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China
| | - Yinan Zhang
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China
| | - Yuhui Shi
- Department of Laboratory Medicine, Fenyang College of Shanxi Medical University, Lvliang, China
| | - Xiaoli Yang
- Reproductive Center, Taiyuan Central Hospital, Taiyuan, China; Reproductive Immunity and Heredity Departments and Cities Jointly Build Key Laboratory Training Bases of Shanxi Province, Taiyuan, China.
| |
Collapse
|
6
|
Skiba MA, Sterling SM, Rawson S, Zhang S, Xu H, Jiang H, Nemeth GR, Gilman MSA, Hurley JD, Shen P, Staus DP, Kim J, McMahon C, Lehtinen MK, Rockman HA, Barth P, Wingler LM, Kruse AC. Antibodies expand the scope of angiotensin receptor pharmacology. Nat Chem Biol 2024; 20:1577-1585. [PMID: 38744986 PMCID: PMC11561159 DOI: 10.1038/s41589-024-01620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
G-protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small-molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue and cellular levels. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays and structural studies, we develop maternally selective heavy-chain-only antibody ('nanobody') antagonists against the angiotensin II type I receptor and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to angiotensin II type I receptor with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.
Collapse
Affiliation(s)
- Meredith A Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sarah M Sterling
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Cryo-EM Facility at MIT.nano, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shuhao Zhang
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Haoran Jiang
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Genevieve R Nemeth
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Morgan S A Gilman
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Joseph D Hurley
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Pengxiang Shen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Dean P Staus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
- Septerna, South San Francisco, CA, USA
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, USA
| | - Conor McMahon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Sanofi, Large Molecule Research, Cambridge, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Laura M Wingler
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Yang Z, Sun T, Wang P, Bai L, Wu Y, Wang T, Li X, Cheng Y, Zhang S, Liu H. The functional subclasses of AT1 receptor autoantibody in patients with coronary heart disease. Biochem Pharmacol 2024; 229:116546. [PMID: 39304102 DOI: 10.1016/j.bcp.2024.116546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/07/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Recently, the identification of autoantibodies (AT1-AA) targeting the second extracellular loop of angiotensin II type 1 receptor (AT1R-ECII) in patients with coronary heart disease (CHD) offers a novel perspective on the interplay between immunity and cardiovascular disease. However, much remains unknown regarding the functional diversity of AT1-AA. In this study, we measured the levels of AT1-AA in the sera of 306 CHD patients and purified AT1-AA from patient's sera (n = 127). The subclasses of AT1-AA were categorized based on their impact on intracellular calcium ([Ca2+]i) levels in mouse arterial smooth muscle cells (MASMCs). Our findings revealed 4 distinct [Ca2+]i response patterns indicating the existence of 4 functional subclasses named H1-, H2-, H3-, and H4-AT1-AA. The correlation analysis demonstrated a positive association between H1-AT1-AA and endogenous coagulation, as well as between H2-AT1-AA and exogenous coagulation; no significant correlation was observed between H3-AT1-AA and the indicators we analyzed. Conversely, H4-AT1-AA exhibited a negative correlation with both leukocyte number and bile acid levels. Logistic regression analysis showed that H2-AT1-AA possessed predictive value for severe CHD. Furthermore, in vitro experiments indicated that both H1- and H2-AT1-AA exerted cytotoxic effects on MASMCs, while H4-AT1-AA increased cell viability. Additionally, an AT1-AA-positive rat model was established by subcutaneously injecting with AT1R-ECII peptide, which produced four similar functional subclasses of rat AT1-AA upon active immunization. This study suggested that classifying different functional subclasses of AT1-AAs can facilitate more accurate evaluation of the condition and prognosis in patients with CHD, thereby providing a novel basis for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Yang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Tao Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Pengli Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Lina Bai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Ye Wu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Tongtong Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Xiaoyan Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China; Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, PR China
| | - Yutong Cheng
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
8
|
Schwartz KS, Stanhewicz AE. Maternal Microvascular Dysfunction During and After Preeclamptic Pregnancy. Compr Physiol 2024; 14:5703-5727. [PMID: 39382165 DOI: 10.1002/cphy.c240003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Preeclampsia, a pregnancy disorder characterized by de novo hypertension and maternal multisystem organ dysfunction, is the leading cause of maternal mortality worldwide and is associated with a fourfold greater risk of cardiovascular disease throughout the lifespan. Current understanding of the etiology of preeclampsia remains unclear, due in part to the varying phenotypical presentations of the disease, which has hindered the development of effective and mechanism-specific treatment or prevention strategies both during and after the affected pregnancy. These maternal sequelae of preeclampsia are symptoms of systemic vascular dysfunction in the maternal nonreproductive microvascular beds that drives the development and progression of adverse cardiovascular outcomes during preeclampsia. Despite normalization of vascular disturbances after delivery, subclinical dysfunction persists in the nonreproductive microvascular beds, contributing to an increased lifetime risk of cardiovascular and metabolic diseases and all-cause mortality. Given that women with a history of preeclampsia demonstrate vascular dysfunction despite an absence of traditional CVD risk factors, an understanding of the underlying mechanisms of microvascular dysfunction during and after preeclampsia is essential to identify potential therapeutic avenues to mitigate or reverse the development of overt disease. This article aims to provide a summary of the existing literature on the pathophysiology of maternal microvascular dysfunction during preeclampsia, the mechanisms underlying the residual dysfunction that remains after delivery, and current and potential treatments both during and after the affected pregnancy that may reduce microvascular dysfunction in these high-risk women. © 2024 American Physiological Society. Compr Physiol 14:5703-5727, 2024.
Collapse
Affiliation(s)
- Kelsey S Schwartz
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, USA
| | - Anna E Stanhewicz
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
9
|
Kokori E, Aderinto N, Olatunji G, Komolafe R, Abraham IC, Babalola AE, Aboje JE, Ukoaka BM, Samuel O, Ayodeji A, Omoworare O, Olatunji D. Maternal and fetal neurocognitive outcomes in preeclampsia and eclampsia; a narrative review of current evidence. Eur J Med Res 2024; 29:470. [PMID: 39342384 PMCID: PMC11437679 DOI: 10.1186/s40001-024-02070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP), such as preeclampsia and eclampsia, present significant risks to maternal and fetal health. While immediate complications are well-documented, emerging research highlights potential neurocognitive impacts on both mothers and their offspring. This narrative review synthesizes evidence on these neurocognitive outcomes associated with HDP, focusing on preeclampsia and eclampsia. A literature search was conducted for studies published from 2000 to February 2024. Maternal outcomes, including memory, executive function, and psychosocial well-being, were assessed across 11 studies, while fetal and neonatal neurocognitive outcomes were explored in five studies. Consistent findings indicate that preeclampsia and eclampsia are linked to impairments in maternal cognitive functions and psychosocial health. Offspring exposed to these conditions in utero also show cognitive deficits and alterations in brain connectivity. Contributing factors include placental dysfunction, altered angiokine levels, maternal stress, and socioeconomic variables. To mitigate these impacts, future research should focus on clarifying the underlying mechanisms and developing early interventions. This review emphasizes the necessity of multidisciplinary approaches to improve neurocognitive outcomes for both mothers and their children affected by preeclampsia and eclampsia.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine, Ladoke Akintola University of Technology, PMB 5000, Ogbomoso, Nigeria.
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Rosemary Komolafe
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | | | - John Ehi Aboje
- College of Health Sciences, Benue State University, Benue, Nigeria
| | | | | | - Akinmeji Ayodeji
- Department of Medicine and Surgery, Olabisi Onabanjo University, Ogun, Nigeria
| | | | - Doyin Olatunji
- Department of Health Sciences, Western Illinois University, Macomb, USA
| |
Collapse
|
10
|
Kermond RF, Kim S, Mackie F, Hahn D, Carroll RP, Sharma A, Durkan AM. Effect of angiotensin II type 1 receptor antibodies on graft function and survival in paediatric kidney transplant recipients. HLA 2024; 104:e15649. [PMID: 39247998 DOI: 10.1111/tan.15649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/25/2024] [Accepted: 08/03/2024] [Indexed: 09/10/2024]
Abstract
HLA donor specific antibodies (DSA) are implicated in antibody-mediated rejection (AMR), graft dysfunction and failure in kidney transplant (KT) recipients. Non-HLA antibodies including angiotensin II type 1 receptor (AT1R) may also play a role in AMR, impact graft function and survival. Data is limited in paediatric KT cohorts. We aimed to assess the prevalence and effect of pre-transplant AT1R antibodies on rejection, graft function and survival in paediatric KT recipients. This was a retrospective cohort study conducted across two paediatric centres including KT recipients with a pre-transplant AT1R antibody level. Outcomes included rejection, de novo DSA formation, graft function, failure, proteinuria and hypertension. Of 71 individuals, 72% recorded a positive pre-transplant AT1R Ab level (≥17 U/mL). Over a median follow-up of 4.7 years, AT1R Ab positivity demonstrated a trend towards increased risk of rejection however was not statistically significant (HR 3.45, 95% CI 0.97-12.35, p-value 0.06). Sensitivity analysis with AT1R Ab levels of ≥25 U/mL (HR 2.05 95% CI 0.78-5.39, p-value 0.14) and ≥40 U/mL (HR 1.32, CI 95% 0.55-3.17, p-value 0.53) validated this. De novo DSA formation occurred more frequently with AT1R Ab positivity (41% vs. 20%, p-value 0.9). AT1R Ab was not associated with hypertension, proteinuria, graft failure or dysfunction. In conclusion, this cohort study demonstrated a high prevalence of pre-transplant AT1R Ab positivity (72%). AT1R Ab positivity demonstrated a trend towards increased risk of rejection and de novo DSA formation however did not meet statistical significance. There was no association between AT1R Ab and hypertension, proteinuria, graft failure or dysfunction.
Collapse
Affiliation(s)
- R F Kermond
- Department of Paediatric Nephrology, Women's and Children's Hospital, Adelaide, Australia
- School of Women's and Children's Health, University of New South Wales, Kensington, Australia
| | - S Kim
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
- Centre for Kidney research, Children's Hospital Westmead, Sydney, Australia
- School of Public Health, Sydney University, Sydney, Australia
| | - F Mackie
- School of Women's and Children's Health, University of New South Wales, Kensington, Australia
- Department of Paediatric Nephrology, Sydney Children's Hospital, Sydney, Australia
| | - D Hahn
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
| | - R P Carroll
- South Australian Transplantation Immunogenetic Laboratory, Australian Red Cross Lifeblood, Adelaide, Australia
- Department of Health Sciences, University of South Australia, Adelaide, Australia
| | - A Sharma
- Department of Nephrology, Westmead Hospital, Sydney, Australia
| | - A M Durkan
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
- School of Paediatrics and Child Health, University of Sydney, Australia
| |
Collapse
|
11
|
Shaw LJ, Patel K, Lala-Trindade A, Feltovich H, Vieira L, Kontorovich A, Ananth C, Taqueti VR, Mitrani L, Stern T, DeBolt C, Kase N, Smith RT, Narula J, Mehran R, Bianco A, Bhatt DL, Stone JL. Pathophysiology of Preeclampsia-Induced Vascular Dysfunction and Implications for Subclinical Myocardial Damage and Heart Failure. JACC. ADVANCES 2024; 3:100980. [PMID: 38938863 PMCID: PMC11198310 DOI: 10.1016/j.jacadv.2024.100980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/04/2024] [Accepted: 02/28/2024] [Indexed: 06/29/2024]
Abstract
Tragically, preeclampsia is a leading cause of pregnancy-related complications and is linked to a heightened risk for morbid and fatal cardiovascular disease (CVD) outcomes. Although the mechanism connecting preeclampsia to CVD risk has yet to be fully elucidated, evidence suggests distinct pathways of early and late preeclampsia with shared CV risk factors but with profound differences in perinatal and postpartum risk to the mother and infant. In early preeclampsia, <34 weeks of gestation, systemic vascular dysfunction contributes to near-term subclinical myocardial damage. Hypertrophy and diastolic abnormalities persist postpartum and contribute to early onset heart failure (HF). This HF risk remains elevated decades later and contributes to premature death. Black women are at the highest risk of preeclampsia and HF. These findings support closer monitoring of women postpartum, especially for those with early and severe preeclampsia to control chronic hypertension and reduce the potentially preventable sequelae of heightened CVD and HF risk.
Collapse
Affiliation(s)
- Leslee J. Shaw
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
- Department of Population Health Science and Policy, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Krishna Patel
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
- Department of Population Health Science and Policy, New York, New York, USA
| | - Anuradha Lala-Trindade
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Helen Feltovich
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Luciana Vieira
- Blavatnik Family Women’s Health Research Institute, New York, New York, USA
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Amy Kontorovich
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Cande Ananth
- Division of Epidemiology and Biostatistics, Department of Obstetrics, Gynecology, and Reproductive Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Viviany R. Taqueti
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsey Mitrani
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Toni Stern
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Chelsea DeBolt
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Nathan Kase
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - R. Theodore Smith
- Department of Opthamology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jagat Narula
- UT Health Houston, McGovern Medical School, Houston, Texas, USA
| | - Roxana Mehran
- Women’s Heart and Vascular Center at Mount Sinai Heart, New York, New York, USA
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Angela Bianco
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| | - Deepak L. Bhatt
- The Lauder Family Cardiovascular Center of Mount Sinai Heart, Samuel Bronfman Department of Medicine (Cardiology), New York, New York, USA
| | - Joanne L. Stone
- Raquel and Jaime Gilinski Department of Obstetrics, Gynecology, and Reproductive Science, New York, New York, USA
| |
Collapse
|
12
|
Hogg JP, Campbell N, Deer E, Fitzgerald S, Cornelius D, Hoang N, Turner T, Amaral L, Lemon JP, Ibrahim T, LaMarca B. The role of T cell stimulated agonistic autoantibodies to the angiotensin II type I receptor (AT1-AA) in mediating multiorgan dysfunction in IL-17 induced hypertension during pregnancy. Am J Reprod Immunol 2024; 91:e13843. [PMID: 38606700 PMCID: PMC11288343 DOI: 10.1111/aji.13843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
PROBLEM Preeclampsia (PE), new-onset hypertension during pregnancy accompanied by organ dysfunction, is associated with chronic inflammation including elevated IL-17, CD4+ T cells, B cells and natural killer (NK) cells. IL-17 can serve as a signal for either the adaptive or innate immune activation. We have previously shown that IL-17 contributes to increased blood pressure in association with elevated TH17 cells, NK cells and B cells secreting angiotensin II type 1 receptor agonistic autoantibodies (AT1-AA) during pregnancy. Moreover, we have shown an important role for CD4+T cells and AT1-AA in multiorgan dysfunction as measured by mitochondrial oxidative stress (mt ROS). However, we do not know the role of adaptive immune cells such as T cells or B cells secreting AT1-AA in mediating the PE phenotype in response to elevated IL-17. METHOD OF STUDY In order to answer this question, we infused IL-17 (150 pg/day i.p.) into either Sprague Dawley (SD) or athymic nude rats via mini-osmotic pump from gestational day (GD) 14-19 of pregnancy. On GD 19, blood pressure was determined and NK cells, mtROS and respiration and AT1-AA production from B cells were measured. RESULTS Infusion of IL-17 increased blood pressure in the presence or absence of T cells. Mean arterial pressure (MAP) increased with IL-17 from 98 ± 2 mm Hg (n = 12) to 114 ± 2 (n = 12) in SD rats and from 99 ± 4 mm Hg (n = 7) versus 115 ± 2 mm Hg (n = 7) in athymic nude rats. Similar trends were seen in NK cells and placental mt ROS. Knowing that IL-17 stimulates AT1-AA in SD pregnant rats, we included a group of SD and athymic nude pregnant rats infused with IL-17 and the AT1-AA inhibitor peptide ('n7AAc'). The inhibitor attenuated blood pressure (104.9 ± 3.2, p = .0001) and normalized NK cells and mt function in SD pregnant rats. Importantly, the AT1-AA was not produced in pregnant nude IL-17 treated rats, nor did 'n7AAc' effect MAP, in nude athymic rats. CONCLUSION These findings suggest two conclusions; one is that IL-17 causes hypertension and multiorgan dysfunction in the absence of T cells and AT1-AA, possibly through its activation of innate cells and secondly, in the presence of T cells, blockade of the AT1-AA attenuates the effect of IL-17. This study indicates the critical effects of elevated IL-17 during pregnancy and suggest treatment modalities to consider for PE women.
Collapse
Affiliation(s)
- James P. Hogg
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, USA
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Denise Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Ngoc Hoang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Ty Turner
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Lorena Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - James P. Lemon
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Babbette LaMarca
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| |
Collapse
|
13
|
Brownfoot F, Rolnik DL. Prevention of preeclampsia. Best Pract Res Clin Obstet Gynaecol 2024; 93:102481. [PMID: 38373378 DOI: 10.1016/j.bpobgyn.2024.102481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/19/2023] [Accepted: 02/05/2024] [Indexed: 02/21/2024]
Abstract
Preeclampsia is a relatively common pregnancy complication and constitutes a major cause of morbidity and mortality for mothers and children worldwide. It disproportionally affects low-resource countries. Appropriate identification of individuals at increased risk and prevention of the disease and its complications remain healthcare and research priorities, and the investigation of potential interventions to prevent preeclampsia has driven much of the obstetric research in recent decades. In this article, we review the scientific literature on the topic, highlighting established benefits and remaining questions regarding different non-pharmacological and pharmacological strategies, including exercise, the timing of birth, aspirin and calcium use, among others, as well as potential novel therapies under investigation.
Collapse
Affiliation(s)
- Fiona Brownfoot
- Mercy Hospital for Women, Heidelberg, Victoria, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel Lorber Rolnik
- Women's and Newborn, Monash Health, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
14
|
Campbell N, Deer E, Solise D, Cornelius DC, Turner T, Amaral LM, Herrock O, Jordan A, Shukla S, Ibrahim T, LaMarca B. AT1-AA Is Produced in Offspring in Response to Placental Ischemia and Is Lowered by B-Cell Depletion Without Compromising Overall Offspring Health. J Am Heart Assoc 2024; 13:e031417. [PMID: 38353227 PMCID: PMC11010106 DOI: 10.1161/jaha.123.031417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/01/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Preeclampsia, new-onset hypertension during pregnancy alongside other organ dysfunction, is the leading cause of mortality for the mother and low birth weight for the baby. Low birth weight contributes to high risk of cardiovascular disorders later in life. Women with preeclampsia have activated B cells producing agonistic autoantibodies to AT1-AA (angiotensin II type I receptor). We hypothesize that rituximab, a B cell-depleting chemotherapeutic, will deplete maternal B cells in reduced uterine perfusion pressure (RUPP) rats without worsening the effect of placental ischemia on pup growth and survival. METHODS AND RESULTS To test this hypothesis, the RUPP procedure was performed, and rituximab was continuously infused via miniosmotic pump. Maternal blood and tissues were collected. A separate group of dams were allowed to deliver, pup weights were recorded, and at 4 months of age, tissues were collected from offspring. Immune cells were measured via flow cytometry, and AT1-AA was quantified using a contraction bioassay. Blood pressure increased in RUPP rats and was normalized with rituximab treatment. RUPP offspring also had increased circulating B cells, cytolytic natural killer cells, and increased circulating AT1-AA, which were normalized with maternal rituximab treatment. This is the first study to analyze the AT1-AA in RUPP offspring, which was normalized with rituximab. CONCLUSIONS Our findings indicate that perinatal rituximab lowers maternal mean arterial pressure in RUPP rats and improves birth weight, circulating AT1-AA, and circulating natural killer cells, indicating that rituximab improves adverse fetal outcomes in response to placental ischemia.
Collapse
Affiliation(s)
- Nathan Campbell
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Evangeline Deer
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Dylan Solise
- Department of Obstetrics and GynecologyUniversity of Mississippi Medical CenterJacksonMS
| | - Denise C. Cornelius
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMS
| | - Ty Turner
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Lorena M. Amaral
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Owen Herrock
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Ariel Jordan
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Shivani Shukla
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Tarek Ibrahim
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Babbette LaMarca
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMS
- Department of Obstetrics and GynecologyUniversity of Mississippi Medical CenterJacksonMS
| |
Collapse
|
15
|
Binda M, Moccaldi B, Civieri G, Cuberli A, Doria A, Tona F, Zanatta E. Autoantibodies Targeting G-Protein-Coupled Receptors: Pathogenetic, Clinical and Therapeutic Implications in Systemic Sclerosis. Int J Mol Sci 2024; 25:2299. [PMID: 38396976 PMCID: PMC10889602 DOI: 10.3390/ijms25042299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Systemic sclerosis (SSc) is a multifaceted connective tissue disease whose aetiology remains largely unknown. Autoimmunity is thought to play a pivotal role in the development of the disease, but the direct pathogenic role of SSc-specific autoantibodies remains to be established. The recent discovery of functional antibodies targeting G-protein-coupled receptors (GPCRs), whose presence has been demonstrated in different autoimmune conditions, has shed some light on SSc pathogenesis. These antibodies bind to GPCRs expressed on immune and non-immune cells as their endogenous ligands, exerting either a stimulatory or inhibitory effect on corresponding intracellular pathways. Growing evidence suggests that, in SSc, the presence of anti-GPCRs antibodies correlates with specific clinical manifestations. Autoantibodies targeting endothelin receptor type A (ETAR) and angiotensin type 1 receptor (AT1R) are associated with severe vasculopathic SSc-related manifestations, while anti-C-X-C motif chemokine receptors (CXCR) antibodies seem to be predictive of interstitial lung involvement; anti-muscarinic-3 acetylcholine receptor (M3R) antibodies have been found in patients with severe gastrointestinal involvement and anti-protease-activated receptor 1 (PAR1) antibodies have been detected in patients experiencing scleroderma renal crisis. This review aims to clarify the potential pathogenetic significance of GPCR-targeting autoantibodies in SSc, focusing on their associations with the different clinical manifestations of scleroderma. An extensive examination of functional autoimmunity targeting GPCRs might provide valuable insights into the underlying pathogenetic mechanisms of SSc, thus enabling the development of novel therapeutic strategies tailored to target GPCR-mediated pathways.
Collapse
Affiliation(s)
- Marco Binda
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Beatrice Moccaldi
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Giovanni Civieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Anna Cuberli
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| | - Francesco Tona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Elisabetta Zanatta
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy; (M.B.)
| |
Collapse
|
16
|
Clark CR, Khalil RA. Regulation of vascular angiotensin II type 1 and type 2 receptor and angiotensin-(1-7)/MasR signaling in normal and hypertensive pregnancy. Biochem Pharmacol 2024; 220:115963. [PMID: 38061417 PMCID: PMC10860599 DOI: 10.1016/j.bcp.2023.115963] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/01/2024]
Abstract
Normal pregnancy (Norm-Preg) is associated with a slight reduction in blood pressure (BP) and decreased BP response to vasoconstrictor stimuli such as angiotensin II (Ang II), although the renin-angiotensin-aldosterone system (RAAS) is upregulated. Preeclampsia (PE) is a complication of pregnancy manifested as hypertension-in-pregnancy (HTN-Preg), and dysregulation of angiotensin biosynthesis and signaling have been implicated. Ang II activates vascular Ang II type-1 receptor (AT1R) and Ang II type-2 receptor (AT2R), while angiotensin-(1-7) promotes Ang-(1-7)/MasR signaling. The role of AT1R in vasoconstriction and the activated cellular mechanisms are well-characterized. The sensitivity of vascular AT1R to Ang II and consequent activation of vasoconstrictor mechanisms decrease during Norm-Preg, but dramatically increase in HTN-Preg. Placental ischemia in late pregnancy could also initiate the release of AT1R agonistic autoantibodies (AT1AA) with significant impact on endothelial dysfunction and activation of contraction pathways in vascular smooth muscle including [Ca2+]c and protein kinase C. On the other hand, the role of AT2R and Ang-(1-7)/MasR in vascular relaxation, particularly during Norm-Preg and PE, is less clear. During Norm-Preg, increases in the expression/activity of vascular AT2R and Ang-(1-7)/MasR promote the production of endothelium-derived relaxing factors such as nitric oxide (NO), prostacyclin and endothelium-derived hyperpolarizing factor leading to generalized vasodilation. Aortic segments of Preg rats show prominent endothelial AT2R staining and increased relaxation and NO production in response to AT2R agonist CGP42112A, and treatment with AT2R antagonist PD123319 enhances phenylephrine-induced contraction. Decreased vascular AT2R and Ang-(1-7)/MasR expression and receptor-mediated mechanisms of vascular relaxation have been suggested in HTN-Preg animal models, but their role in human PE needs further testing. Changes in angiotensin-converting enzyme-2 (ACE2) have been observed in COVID-19 patients, and whether ACE2 influences the course of COVID-19 viral infection/immunity in Norm-Preg and PE is an intriguing area for research.
Collapse
Affiliation(s)
- Caroline R Clark
- 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
|
17
|
Lin B, Robinson L, Soliman B, Gulizia J, Usala S. Autoimmune Implications in a Patient with Graves' Hyperthyroidism, Pre-eclampsia with Severe Features, and Primary Aldosteronism. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:170. [PMID: 38256430 PMCID: PMC10820415 DOI: 10.3390/medicina60010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024]
Abstract
Background and Objectives: Graves' disease (GD) and primary aldosteronism (PA) are two pathologies that can cause significant morbidity and mortality. GD is mediated by autoantibodies, and recent studies have shown autoantibody involvement in the pathophysiology behind both PA and pre-eclampsia. The coexistence of GD and PA, however, is reportedly rare. This report describes a unique case of Graves' hyperthyroidism and concomitant PA in a patient with a history of pre-eclampsia with severe features. Case Presentation: The patient presented at 17 weeks pregnancy with mild hyperthyroidism, negative TSH receptor antibodies, and a low level of thyroid-stimulating immunoglobulins (TSI). Her TSH became detectable with normal thyroid hormone levels, and therefore, no anti-thyroid medication was administered. At 34 weeks she developed pre-eclampsia with severe features, and a healthy child was delivered; her TSH returned to normal. Seven months after delivery, she presented emergently with severe hyperthyroidism, hypertensive crisis, and a serum potassium of 2.5 mmol/L. Her hypertension was uncontrolled on multiple anti-hypertensives. Both TSI and TSH receptor antibodies were negative. The aldosterone(ng/dL)/renin(ng/mL/h ratio was (13/0.06) = 216.7, and abdominal CT imaging demonstrated normal adrenal glands; thus, a diagnosis of PA was made. Her blood pressure was subsequently controlled with only spironolactone at 50 mg 2xday. Methimazole was started but discontinued because of an allergic reaction. Consequently, a thyroidectomy was performed, and pathology revealed Graves' disease. The patient remained well on levothyroxine at 125 mcg/day and spironolactone at 50 mg 2xday three months after the thyroidectomy. Conclusions: This patient manifested severe GD with antibodies undetectable by conventional TSI and TSH receptor assays and accelerated hypertension from PA simultaneously. These conditions were successfully treated separately by spironolactone and thyroidectomy. Autoimmune PA was considered likely given the clinical picture. The diagnosis of PA should be considered in hypertension with GD.
Collapse
Affiliation(s)
- Benjamin Lin
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.L.); (L.R.)
| | - Lauren Robinson
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.L.); (L.R.)
| | - Basem Soliman
- Department of Surgery, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA;
| | - Jill Gulizia
- Women’s Healthcare Associates, Obstetrics and Gynecology, Amarillo, TX 79106, USA;
| | - Stephen Usala
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
18
|
Zhang X, Li B, Yan Y, Sun F, Zhang S, Wang M, Liu H. AT1R autoantibody promotes phenotypic transition of smooth muscle cells by activating AT1R-OAS2. Biochem Pharmacol 2024; 219:115977. [PMID: 38092283 DOI: 10.1016/j.bcp.2023.115977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic transition of vascular smooth muscle cells (VSMCs) is an early event in the onset and progression of several cardiovascular diseases. As an important mediator of the renin-angiotensin system (RAS), activation of the angiotensin II type 1 receptor (AT1R) induces phenotypic transition of VSMCs. AT1R autoantibodies (AT1-AAs), which are agonistic autoantibodies of AT1R, have been detected in the sera of patients with a variety of cardiovascular diseases associated with phenotypic transition. However, the effect of AT1-AA on phenotypic transition is currently unknown. In this study, AT1-AA-positive rat model was established by active immunization to detect markers of VSMCs phenotypic transition. The results showed that AT1-AA-positive rats showed phenotypic transition of VSMCs, which was evidenced by the decrease of contractile markers, while the increase of synthetic markers in the thoracic aorta. However, in AT1-AA-positive AT1R knockout rats, the phenotypic transition-related proteins were not altered. In vitro, after stimulating human aortic smooth muscle cells with AT1-AA for 48 h, 2'-5' oligoadenylate synthase 2 (OAS2) was identified as the key differentially expressed gene by RNA sequencing and bioinformatics analysis. Furthermore, high expression of OAS2 was found in aorta of AT1-AA-positive rats; knockdown of OAS2 by siRNA can reverse the phenotypic transition of VSMCs induced by AT1-AA. In summary, this study suggests that AT1-AA can promote phenotypic transition of VSMCs through AT1R-OAS2 pathway, and OAS2 might serve as a potential therapeutic target to prevent pathological phenotypic transition of smooth muscle cells.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Bingjie Li
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yingshuo Yan
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Fei Sun
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
19
|
Liman TG, Siegerink B, Piper S, Catar R, Moll G, Riemekasten G, Heidecke H, Heuschmann PU, Elkind MSV, Dragun D, Endres M. Vasoregulatory Autoantibodies and Clinical Outcome After Ischemic Stroke-PROSCIS-B. J Am Heart Assoc 2023; 12:e032441. [PMID: 38014691 PMCID: PMC10727313 DOI: 10.1161/jaha.123.032441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Vasoregulatory autoantibodies including autoantibodies targeting G-protein-coupled receptors might play a functional role in vascular diseases. We investigated the impact of vasoregulatory autoantibodies on clinical outcome after ischemic stroke. METHODS AND RESULTS Data were used from the PROSCIS-B (Prospective Cohort With Incident Stroke-Berlin). Autoantibody-targeting receptors such as angiotensin II type 1 receptor (AT1R), endothelin-1 type A receptor, complement factor-3 and -5 receptors, vascular endothelial growth factor receptor-1 and -2, vascular endothelial growth factor A and factor B were measured. We explored associations of high antibody levels with (1) poor functional outcome defined as modified Rankin Scale >2 or Barthel Index <60 at 1 year after stroke, (2) Barthel Index scores over time using general estimating equations, and (3) secondary vascular events (recurrent stroke, myocardial infarction) or death up to 3 years using Cox proportional hazard models. We included 491 patients with ischemic stroke with data on autoantibody levels and outcome. In models adjusted for demographics and vascular risk factors, high autoantibody concentrations (quartile 4) targeting complement factor C3a receptor, vascular endothelial growth factor receptor-2, and vascular endothelial growth factor B were associated with poor functional outcome at 1 year: (odds ratio, 2.0 [95% CI, 1.1-3.6]; odds ratio, 1.8 [95% CI, 1.1-3.2]; and odds ratio, 2.1 [95% CI, 1.2-3.6], respectively) and with lower Barthel Index scores over 3 years (complement factor C3a receptor: adjusted β=-3.3 [95% CI, -5.7 to -0.5]; VEGF-B: adjusted β=-2.4 [95% CI, -4.8 to -0.06]). Patients with high autoantibody levels were not at higher risk for secondary vascular events or death. CONCLUSIONS High levels of autoantibodies against vascular endothelial growth factor receptor-2, vascular endothelial growth factor B, and complement factor C3a receptor measured are associated with poor functional outcome after stroke but not with recurrent vascular events or death. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT01363856.
Collapse
Affiliation(s)
- Thomas G. Liman
- Center for Stroke Research Berlin (CSB)Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Neurology at Evangelical Hospital OldenburgCarl von Ossietzky UniversityOldenburgGermany
- German Center for Neurodegenerative Diseases (DZNE)Site BerlinGermany
| | - Bob Siegerink
- Leiden University Medical Center, Leiden UniversityDepartment of Clinical EpidemiologyLeidenThe Netherlands
| | - Sophie Piper
- Institute for Biometry and Clinical Epidemiology; Charité – Universitätsmedizin BerlinBerlinGermany
| | - Rusan Catar
- Department of Nephrology and Intensive Care MedicineCharité – Universitätsmedizin BerlinBerlinGermany
| | - Guido Moll
- Department of Nephrology and Intensive Care MedicineCharité – Universitätsmedizin BerlinBerlinGermany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical ImmunologyUniversitätsklinikum Schleswig‐HolsteinLübeckGermany
| | | | - Peter U. Heuschmann
- Comprehensive Heart Failure CenterUniversity Hospital WürzburgWürzburgGermany
- Institute of Clinical Epidemiology and Biometry, University of WürzburgWürzburgGermany
| | - Mitchell S. V. Elkind
- Department of Neurology, Vagelos College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public HealthColumbia UniversityNew YorkNYUSA
| | - Duska Dragun
- Department of Nephrology and Intensive Care MedicineCharité – Universitätsmedizin BerlinBerlinGermany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB)Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Neurology with Experimental NeurologyCharité – Universitätsmedizin BerlinBerlinGermany
- German Center for Neurodegenerative Diseases (DZNE)Site BerlinGermany
- German Centre for Cardiovascular Research (DZHK)BerlinGermany
| |
Collapse
|
20
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
21
|
Skiba MA, Sterling SM, Rawson S, Gilman MS, Xu H, Nemeth GR, Hurley JD, Shen P, Staus DP, Kim J, McMahon C, Lehtinen MK, Wingler LM, Kruse AC. Antibodies Expand the Scope of Angiotensin Receptor Pharmacology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.23.554128. [PMID: 37662341 PMCID: PMC10473732 DOI: 10.1101/2023.08.23.554128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
G protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue, and cellular level. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays, and structural studies, we develop maternally selective heavy chain-only antibody ("nanobody") antagonists against the angiotensin II type I receptor (AT1R) and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to AT1R with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.
Collapse
Affiliation(s)
- Meredith A. Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah M. Sterling
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Morgan S.A. Gilman
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Huixin Xu
- Department of Pathology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Genevieve R. Nemeth
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph D. Hurley
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Pengxiang Shen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Dean P. Staus
- Department of Medicine and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Jihee Kim
- Department of Medicine and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Conor McMahon
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Maria K. Lehtinen
- Department of Pathology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Laura M. Wingler
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Andrew C. Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Campbell N, Solise D, Deer E, LaMarca B. Sex Differences in Offspring of Preeclamptic Pregnancies. CURRENT OPINION IN PHYSIOLOGY 2023; 34:100688. [PMID: 37305157 PMCID: PMC10249590 DOI: 10.1016/j.cophys.2023.100688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A poor uterine environment causes changes in fetal development that affect the health of offspring long-term. Although there are multiple pathways that contribute to the development of cardiovascular and neurological disease, low birth weight or fetal growth restriction (FGR) predisposes offspring to these diseases. There is a link between fetal exposure to adverse influences and hypertension later in life. Many epidemiological studies support the link between fetal life and the risk of disease later in life. Experimental models have sought to provide mechanistic proof of this link while simultaneously investigating potential therapeutics or treatment pathways. Preeclampsia (PE), one of several hypertensive disorders in pregnancy, is a leading cause of morbidity and mortality for both the mother and fetus. Studies have shown that PE is a state of chronic inflammation and there is an imbalance between pro-inflammatory and regulatory immune cells and mediators. There is no cure for PE beyond the delivery of the fetal-placental unit, and many PE pregnancies result in FGR and preterm birth. Epidemiological data demonstrate that the sex of the offspring is correlated with the degree of cardiovascular disease that develops with the age of the offspring yet few studies examine the effect of sex on the development of neurological disorders. Even fewer studies examine the effects of therapeutics on offspring of different genders following a PE pregnancy. Moreover, there remain significant gaps in knowledge concerning the role the immune system plays in FGR offspring developing hypertension or neurovascular disorders later in life. Therefore, the purpose of this review is to highlight current research on sex differences in the developmental programming of hypertension and neurological disorders following a PE pregnancy.
Collapse
Affiliation(s)
- Nathan Campbell
- Department of Pharmacology & Toxicology, University of
Mississippi Medical Center, Jackson, MS
| | - Dylan Solise
- Department of Obstetrics and Gynecology, University of
Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of
Mississippi Medical Center, Jackson, MS
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of
Mississippi Medical Center, Jackson, MS
- Department of Obstetrics and Gynecology, University of
Mississippi Medical Center, Jackson, MS
| |
Collapse
|
23
|
Kaltsas A, Zikopoulos A, Moustakli E, Zachariou A, Tsirka G, Tsiampali C, Palapela N, Sofikitis N, Dimitriadis F. The Silent Threat to Women's Fertility: Uncovering the Devastating Effects of Oxidative Stress. Antioxidants (Basel) 2023; 12:1490. [PMID: 37627485 PMCID: PMC10451552 DOI: 10.3390/antiox12081490] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Oxidative stress (OS), which arises through an imbalance between the formation of reactive oxygen species (ROS) and antioxidant defenses, plays a key role in the pathophysiology of female infertility, with the latter constituting just one of a number of diseases linked to OS as a potential cause. The aim of the present article is to review the literature regarding the association between OS and female infertility. Among the reproductive diseases considered are endometriosis and polycystic ovary syndrome (PCOS), while environmental pollutants, lifestyle variables, and underlying medical conditions possibly resulting in OS are additionally examined. Current evidence points to OS likely contributing to the pathophysiology of the above reproductive disorders, with the amount of damage done by OS being influenced by such variables as duration and severity of exposure and the individual's age and genetic predisposition. Also discussed are the processes via which OS may affect female fertility, these including DNA damage and mitochondrial dysfunction. Finally, the last section of the manuscript contains an evaluation of treatment options, including antioxidants and lifestyle modification, capable of minimizing OS in infertile women. The prime message underlined by this review is the importance of considering OS in the diagnosis and treatment of female infertility. Further studies are, nevertheless required to identify the best treatment regimen and its ideal duration.
Collapse
Affiliation(s)
- Aris Kaltsas
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Athanasios Zikopoulos
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Efthalia Moustakli
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.); (G.T.)
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Georgia Tsirka
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.); (G.T.)
| | | | - Natalia Palapela
- Medical Faculty, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
24
|
Tabacco S, Ambrosii S, Polsinelli V, Fantasia I, D’Alfonso A, Ludovisi M, Cecconi S, Guido M. Pre-Eclampsia: From Etiology and Molecular Mechanisms to Clinical Tools-A Review of the Literature. Curr Issues Mol Biol 2023; 45:6202-6215. [PMID: 37623210 PMCID: PMC10453909 DOI: 10.3390/cimb45080391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
Pre-eclampsia is a severe pregnancy-related complication that manifests as a syndrome with multisystem involvement and damage. It has significantly grown in frequency during the past 30 years and could be considered as one of the major causes of maternal and fetal morbidity and mortality. However, the specific etiology and molecular mechanisms of pre-eclampsia are still poorly known and could have a variety of causes, such as altered angiogenesis, inflammations, maternal infections, obesity, metabolic disorders, gestational diabetes, and autoimmune diseases. Perhaps the most promising area under investigation is the imbalance of maternal angiogenic factors and its effects on vascular function, though studies in placental oxidative stress and maternal immune response have demonstrated intriguing findings. However, to determine the relative importance of each cause and the impact of actions aiming to significantly reduce the incidence of this illness, more research is needed. Moreover, it is necessary to better understand the etiologies of each subtype of pre-eclampsia as well as the pathophysiology of other major obstetrical syndromes to identify a clinical tool able to recognize patients at risk of pre-eclampsia early.
Collapse
Affiliation(s)
- Sara Tabacco
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Silvia Ambrosii
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Valentina Polsinelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Ilaria Fantasia
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Angela D’Alfonso
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Manuela Ludovisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Maurizio Guido
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
25
|
Campbell N, Strong L, Fang X, Border JJ, Herrock O, Turner T, Deer E, Amaral L, Dechend R, Roman RJ, LaMarca B. AT1-AA Infusion during Pregnancy Impairs CBF Autoregulation Postpartum. INTERNATIONAL JOURNAL OF CEREBROVASCULAR DISEASE AND STROKE 2023; 6:154. [PMID: 37901747 PMCID: PMC10610033 DOI: 10.29011/2688-8734.100154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Preeclampsia (PE), new-onset hypertension during pregnancy alongside organ dysfunction, is a leading cause of morbidity and mortality for the mother and fetus. PE women have activated B cells that produce agonistic autoantibodies to the angiotensin II type 1 receptor (AT1-AA). AT1-AA impairs cerebral blood flow (CBF) autoregulation during pregnancy. Although AT1-AA often remains elevated up to 8 years postpartum, AT1-AA's effect on CBF autoregulation postpartum is unknown. This study examined whether elevated AT1-AA during pregnancy impairs CBF autoregulation postpartum and if this was augmented by infusion of AT1-AA postpartum. AT1-AA was infused into 12-week-old timed-pregnant Sprague Dawley rats beginning on gestational day 14. Uterine artery resistance index (UARI) was measured on gestational day 18 as a measure of endothelial dysfunction associated with PE. Dams were allowed to deliver. One group was given a second infusion of AT1-AA (50% perinatal dose mimicking levels observed in postpartum PE women) at 9 weeks postpartum. After postpartum week 10, mean arterial pressure (MAP) was measured in conscious rats and CBF autoregulation was measured by laser Doppler flowmetry. AT1-AA during pregnancy increased UARI (P<0.05). AT1-AA during pregnancy did not affect MAP postpartum but did impair CBF autoregulation postpartum. Infusion of AT1-AA postpartum significantly elevated blood pressure (P<0.01) but did not further impair CBF autoregulation. This study demonstrates that circulating AT1-AA during pregnancy causes impairment of CBF autoregulation well into the postpartum period indicating that elevated AT1-AA leads to long-term cerebrovascular consequences. Targeting AT1-AA may prevent cerebrovascular effects associated with PE during pregnancy and postpartum.
Collapse
Affiliation(s)
- Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Luke Strong
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xing Fang
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jane J Border
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lorena Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ralf Dechend
- Charité, Campus Buch, Experimental and Clinical Research Center, HELIOS Clinic, Berlin-Buch, Germany
| | - Richard J Roman
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
26
|
Zhang X, Zhang S, Wang M, Chen H, Liu H. Advances in the allostery of angiotensin II type 1 receptor. Cell Biosci 2023; 13:110. [PMID: 37330563 DOI: 10.1186/s13578-023-01063-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
Angiotensin II type 1 receptor (AT1R) is a promising therapeutic target for cardiovascular diseases. Compared with orthosteric ligands, allosteric modulators attract considerable attention for drug development due to their unique advantages of high selectivity and safety. However, no allosteric modulators of AT1R have been applied in clinical trials up to now. Except for the classical allosteric modulators of AT1R such as antibody, peptides and amino acids, cholesterol and biased allosteric modulators, there are non-classical allosteric modes including the ligand-independent allosteric mode, and allosteric mode of biased agonists and dimers. In addition, finding the allosteric pockets based on AT1R conformational change and interaction interface of dimers are the future of drug design. In this review, we summarize the different allosteric mode of AT1R, with a view to contribute to the development and utilization of drugs targeting AT1R allostery.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hao Chen
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, People's Republic of China.
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao, You An Men Street, Beijing, 100069, China.
| |
Collapse
|
27
|
Darka Aslan I, Sel G, Barut F, Baser Acikgoz R, Balci S, Ozmen U, Barut A, Harma M, Harma MI. Investigation of CD56, ADAM17 and FGF21 Expressions in the Placentas of Preeclampsia Cases. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1145. [PMID: 37374349 DOI: 10.3390/medicina59061145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023]
Abstract
Objective: In the present study, we investigated the expression of CD56, ADAM17 and FGF21 antibodies (Ab), which we think have an effect on the pathophysiology of preeclampsia (PE), in pregnant patients with healthy placentas and placentas with PE. The expression of these antibodies has been investigated in a limited amount of former research, but their role in PE has not yet been clarified. With this study, we aimed to contribute to the elucidation of the pathophysiology of PE and the detection of new target molecules for treatment. Materials and Methods: Parturients with singleton pregnancy at 32 weeks or above without any maternal or fetal pathology who were admitted to the Department of Obstetrics and Gynecology, Zonguldak Bülent Ecevit University Practice and Research Hospital between 11 January 2020 and 7 January 2022 were included in the present study. Pregnant women with coexisting disease or a pathology related to the placenta (ablation placenta, vasa previa, hemangioma, etc.) were excluded. CD56, ADAM17 and FGF21 antibodies were histopathologically and immunohistochemically detected in 60 placentas with PE (study group) and 43 healthy placentas (control group). Results: CD56, ADAM17 and FGF21 proteins were all more intensely expressed in preeclamptic placentas and a statistically significant difference was found between the two groups for all three antibodies (p < 0.001). Deciduitis, perivillous fibrin deposition, intervillous fibrin, intervillous hemorrhage, infarct, calcification, laminar necrosis and syncytial node were found to be significantly more common in the study group (p < 0.001). Conclusions: We observed that CD56, ADAM17 and FGF21 expressions increased in preeclamptic placentas. These Ab may be responsible for the pathogenesis of PE, which can be illuminated with further studies.
Collapse
Affiliation(s)
- Irem Darka Aslan
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Gorker Sel
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Figen Barut
- Department of Medical Pathology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Rabia Baser Acikgoz
- Department of Gynecology and Obsterics, Zonguldak Alapli Public Hospital, Yeni Siteler Street, 67850 Zonguldak, Turkey
| | - Sibel Balci
- Department of Biostatistics, Faculty of Medicine, Kocaeli University, 41380 Kocaeli, Turkey
| | - Ulku Ozmen
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Aykut Barut
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Muge Harma
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| | - Mehmet Ibrahim Harma
- Department of Gynecology and Obsterics, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenköy, Kozlu, 67000 Zonguldak, Turkey
| |
Collapse
|
28
|
Wang X, Shields CA, Ekperikpe U, Amaral LM, Williams JM, Cornelius DC. VASCULAR AND RENAL MECHANISMS OF PREECLAMPSIA. CURRENT OPINION IN PHYSIOLOGY 2023; 33:100655. [PMID: 37009057 PMCID: PMC10062189 DOI: 10.1016/j.cophys.2023.100655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Preeclampsia (PE) is a multisystem obstetric disorder that affects 2-10% of pregnancies worldwide and it is a leading cause of maternal and fetal morbidity and mortality. The etiology of PE development is not clearly delineated, but since delivery of the fetus and placenta often leads to symptom resolution in the most cases of PE, it is hypothesized that the placenta is the inciting factor of the disease. Current management strategies for PE focus on treating the maternal symptoms to stabilize the mother in an attempt to prolong the pregnancy. However, the efficacy of this management strategy is limited. Therefore, identification of novel therapeutic targets and strategies is needed. Here, we provide a comprehensive overview of the current state of knowledge regarding mechanisms of vascular and renal pathophysiology during PE and discuss potential therapeutic targets directed at improving maternal vascular and renal function.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology, University of Mississippi Medical Center
| | - Corbin A Shields
- Department of Emergency Medicine, University of Mississippi Medical Center
| | - Ubong Ekperikpe
- Department of Pharmacology, University of Mississippi Medical Center
| | - Lorena M Amaral
- Department of Pharmacology, University of Mississippi Medical Center
| | | | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center
- Department of Emergency Medicine, University of Mississippi Medical Center
| |
Collapse
|
29
|
Solise D, Campbell N, Ashraf U, Herrock O, Crudup B, Mallette J, Willis A, Rawls AZ, Turner T, Cockrell K, Zheng B, Deer E, Amaral L, Alexander BT, Lamarca B. Inhibition of angiotensin II type 1 receptor agonistic autoantibodies by direct binding does not impact reduced uterine perfusion pressure offspring birthweight and blood pressure at adulthood. Am J Obstet Gynecol MFM 2023; 5:100945. [PMID: 36990181 PMCID: PMC10449034 DOI: 10.1016/j.ajogmf.2023.100945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Preeclampsia, a new-onset hypertension with end-organ damage in pregnancy, is associated with maternal death and morbidity, low birthweight, and B cells producing agonistic autoantibodies to the angiotensin II type 1 receptor. Angiotensin II type 1 receptor agonistic autoantibodies are produced during pregnancy and after delivery and are in the fetal circulation of women with preeclampsia. Angiotensin II type 1 receptor agonistic autoantibodies are shown to contribute to endothelial dysfunction, renal dysfunction, hypertension, fetal growth restriction, and chronic inflammation in women with preeclampsia. The reduced uterine perfusion pressure rat model of preeclampsia exhibits these features. In addition, we have shown that the administration of a 'n7AAc', which blocks the actions of the angiotensin II type 1 receptor autoantibodies, improves preeclamptic features in the rat with reduced uterine perfusion pressure. However, the effect of a 'n7AAc' on the long-term health of the offspring of rats with reduced uterine perfusion pressure is unknown. OBJECTIVE This study aimed to test the hypothesis that inhibition of angiotensin II type 1 receptor autoantibodies during pregnancy will improve offspring birthweight and prevent increased cardiovascular risk in offspring in adulthood. STUDY DESIGN To test our hypothesis, a 'n7AAc' (24 µg/d) or vehicle (saline) was given on gestation day 14 via miniosmotic pumps to sham-operated (sham) and Sprague-Dawley rat dams with reduced uterine perfusion pressure. Dams were allowed to deliver naturally, and pup weights were recorded within 12 hours after birth. Pups were aged to 16 weeks, at which time mean arterial pressure was measured and whole blood was collected to measure immune cells by flow cytometry, cytokines by enzyme-linked immunosorbent assay, and angiotensin II type 1 receptor autoantibodies by bioassay. A 2-way analysis of variance with the Bonferroni multiple comparison posthoc test was used for statistical analysis. RESULTS There was no significant change in offspring birthweight of 'n7AAc'-treated male (5.63±0.09 g) or female (5.66±0.14 g) offspring from reduced uterine perfusion pressure dams compared with vehicle male (5.51±0.17 g) or female (5.74±0.13 g) offspring from reduced uterine perfusion pressure dams. In addition, 'n7AAc' treatment did not affect the birthweight of sham male (5.83±0.11 g) or female (5.64±0.12) offspring compared with vehicle sham male (5.811±0.15 g) or female (5.40±0.24 g) offspring. At adulthood, mean arterial pressure was unchanged in 'n7AAc' treated-male (133±2 mm Hg) and female (127±3 mm Hg) offspring from reduced uterine perfusion pressure dams compared with vehicle male (142±3 mm Hg) and female (133±5 mm Hg) offspring from reduced uterine perfusion pressure dams, the 'n7AAc'-treated sham male (133±3 mm Hg) and female (135±3 mm Hg) offspring, and vehicle sham male (138±4 mm Hg) and female (130±5 mm Hg) offspring. The circulating angiotensin II type 1 receptor autoantibodies were increased in vehicle male (10±2 ΔBPM) and female (14±2 ΔBPM) offspring from reduced uterine perfusion pressure dams and 'n7AAc'-treated male (11±2 ΔBPM) and female (11±2 ΔBPM) offspring from reduced uterine perfusion pressure dams compared with vehicle sham male (1±1 ΔBPM) and female (-1±1 ΔBPM) offspring and 'n7AAc'-treated sham male (-2±2 ΔBPM) and female (-2±2 ΔBPM) offspring. CONCLUSION Our findings indicated that perinatal 7-amino acid sequence peptide treatment does not negatively impact offspring survival or weight at birth. Perinatal 'n7AAc' treatment did not prevent increased cardiovascular risk in offspring, but it also did not cause an increased cardiovascular risk in offspring with reduced uterine perfusion pressure compared with controls. Furthermore, perinatal 'n7AAc' treatment did not affect endogenous immunologic programming as observed by no change in circulating angiotensin II type 1 receptor autoantibodies in either sex of adult offspring from reduced uterine perfusion pressure dams.
Collapse
Affiliation(s)
- Dylan Solise
- Department of Obstetrics and Gynecology (Drs Solise and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Nathan Campbell
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Usman Ashraf
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Owen Herrock
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Breland Crudup
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Jordan Mallette
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Alex Willis
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Adam Z Rawls
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Ty Turner
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Kathy Cockrell
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Baoying Zheng
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Lorena Amaral
- Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS
| | - Barbara T Alexander
- Department of Physiology and Biophysics (Drs Ashraf and Crudup, Ms Mallette, Messrs Willis and Rawls, Ms Cockrell, and Dr Alexander), University of Mississippi Medical Center, Jackson, MS
| | - Babbette Lamarca
- Department of Obstetrics and Gynecology (Drs Solise and Lamarca), University of Mississippi Medical Center, Jackson, MS; Department of Pharmacology and Toxicology (Messrs Campbell, Herrock, Turner, and Zheng and Drs Deer, Amaral, and Lamarca), University of Mississippi Medical Center, Jackson, MS.
| |
Collapse
|
30
|
Alexander BT, South AM, August P, Bertagnolli M, Ferranti EP, Grobe JL, Jones EJ, Loria AS, Safdar B, Sequeira-Lopez MLS. Appraising the Preclinical Evidence of the Role of the Renin-Angiotensin-Aldosterone System in Antenatal Programming of Maternal and Offspring Cardiovascular Health Across the Life Course: Moving the Field Forward: A Scientific Statement From the American Heart Association. Hypertension 2023; 80:e75-e89. [PMID: 36951054 PMCID: PMC10242542 DOI: 10.1161/hyp.0000000000000227] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
There is increasing interest in the long-term cardiovascular health of women with complicated pregnancies and their affected offspring. Emerging antenatal risk factors such as preeclampsia appear to increase the risk of hypertension and cardiovascular disease across the life course in both the offspring and women after pregnancy. However, the antenatal programming mechanisms responsible are complex and incompletely understood, with roots in alterations in the development, structure, and function of the kidney, heart, vasculature, and brain. The renin-angiotensin-aldosterone system is a major regulator of maternal-fetal health through the placental interface, as well as kidney and cardiovascular tissue development and function. Renin-angiotensin-aldosterone system dysregulation plays a critical role in the development of pregnancy complications such as preeclampsia and programming of long-term adverse cardiovascular health in both the mother and the offspring. An improved understanding of antenatal renin-angiotensin-aldosterone system programming is crucial to identify at-risk individuals and to facilitate development of novel therapies to prevent and treat disease across the life course. Given the inherent complexities of the renin-angiotensin-aldosterone system, it is imperative that preclinical and translational research studies adhere to best practices to accurately and rigorously measure components of the renin-angiotensin-aldosterone system. This comprehensive synthesis of preclinical and translational scientific evidence of the mechanistic role of the renin-angiotensin-aldosterone system in antenatal programming of hypertension and cardiovascular disease will help (1) to ensure that future research uses best research practices, (2) to identify pressing needs, and (3) to guide future investigations to maximize potential outcomes. This will facilitate more rapid and efficient translation to clinical care and improve health outcomes.
Collapse
|
31
|
Deer E, Herrock O, Campbell N, Cornelius D, Fitzgerald S, Amaral LM, LaMarca B. The role of immune cells and mediators in preeclampsia. Nat Rev Nephrol 2023; 19:257-270. [PMID: 36635411 PMCID: PMC10038936 DOI: 10.1038/s41581-022-00670-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 01/14/2023]
Abstract
Preeclampsia is a hypertensive disorder of major concern in pregnancy than can lead to intrauterine growth restriction, placental abruption and stillbirth. The pathophysiology of preeclampsia is multifactorial, including not only kidney dysfunction but also endothelial dysfunction, as the maternal endothelium becomes exposed to placental factors that are released into the circulation and increase systemic levels of vasoconstrictors, oxidative stress, anti-angiogenic factors and inflammatory mediators. Importantly, inflammation can lead to insufficient placental perfusion and low birthweight in offspring. Various innate and adaptive immune cells and mediators have been implicated in the development of preeclampsia, in which oxidative stress is associated with activation of the maternal inflammatory response. Immune cells such as regulatory T cells, macrophages, natural killer cells, and neutrophils are known to have major causative roles in the pathology of preeclampsia, but the contributions of additional immune cells such as B cells, inflammatory cytokines and anti-angiotensin II type 1 receptor autoantibodies are also now recognized. Immunological interventions, therefore, have therapeutic potential in this disease. Here, we provide an overview of the immune responses that are involved in the pathogenesis of preeclampsia, including the role of innate and adaptive immune cells and mediators.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Denise Cornelius
- Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sarah Fitzgerald
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
32
|
Mallette JH, Crudup BF, Alexander BT. Growth Restriction in Preeclampsia: Lessons from Animal Models. CURRENT OPINION IN PHYSIOLOGY 2023; 32:100647. [PMID: 36968132 PMCID: PMC10035651 DOI: 10.1016/j.cophys.2023.100647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Preeclampsia remains a major health concern for mother and child. Yet, treatment options remain limited to early delivery. Placental dysfunction in preeclampsia occurs in response to an increase in oxidative stress and inflammatory cytokines with vasoactive and anti-angiogenic factors contributing to impaired maternal and fetal health. Moreover, recent studies indicate a potential role for epigenetic mediators in the pathophysiology of placental ischemia. Numerous animal models are utilized to explore the pathogenesis of preeclampsia and fetal growth restriction. This review provides a brief overview of recent progress in preclinical studies regarding potential therapeutic targets for the treatment and prevention of preeclampsia with an emphasis on fetal growth restriction and the fetal programming of increased cardiovascular risk.
Collapse
Affiliation(s)
- Jordan H. Mallette
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS USA
| | - Breland F. Crudup
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS USA
| | - Barbara T. Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS USA
| |
Collapse
|
33
|
Wang J, Zhang P, Liu M, Huang Z, Yang X, Ding Y, Liu J, Cheng X, Xu S, He M, Zhang F, Wang G, Li R, Yang X. Alpha-2-macroglobulin is involved in the occurrence of early-onset pre-eclampsia via its negative impact on uterine spiral artery remodeling and placental angiogenesis. BMC Med 2023; 21:90. [PMID: 36894970 PMCID: PMC9999529 DOI: 10.1186/s12916-023-02807-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Pre-eclampsia (PE) is one of the leading causes of maternal and fetal morbidity/mortality during pregnancy, and alpha-2-macroglobulin (A2M) is associated with inflammatory signaling; however, the pathophysiological mechanism by which A2M is involved in PE development is not yet understood. METHODS Human placenta samples, serum, and corresponding clinical data of the participants were collected to study the pathophysiologic mechanism underlying PE. Pregnant Sprague-Dawley rats were intravenously injected with an adenovirus vector carrying A2M via the tail vein on gestational day (GD) 8.5. Human umbilical artery smooth muscle cells (HUASMCs), human umbilical vein endothelial cells (HUVECs), and HTR-8/SVneo cells were transfected with A2M-expressing adenovirus vectors. RESULTS In this study, we demonstrated that A2M levels were significantly increased in PE patient serum, uterine spiral arteries, and feto-placental vasculature. The A2M-overexpression rat model closely mimicked the characteristics of PE (i.e., hypertension in mid-to-late gestation, histological and ultrastructural signs of renal damage, proteinuria, and fetal growth restriction). Compared to the normal group, A2M overexpression significantly enhanced uterine artery vascular resistance and impaired uterine spiral artery remodeling in both pregnant women with early-onset PE and in pregnant rats. We found that A2M overexpression was positively associated with HUASMC proliferation and negatively correlated with cell apoptosis. In addition, the results demonstrated that transforming growth factor beta 1 (TGFβ1) signaling regulated the effects of A2M on vascular muscle cell proliferation described above. Meanwhile, A2M overexpression regressed rat placental vascularization and reduced the expression of angiogenesis-related genes. In addition, A2M overexpression reduced HUVEC migration, filopodia number/length, and tube formation. Furthermore, HIF-1α expression was positively related to A2M, and the secretion of sFLT-1 and PIGF of placental origin was closely related to PE during pregnancy or A2M overexpression in rats. CONCLUSIONS Our data showed that gestational A2M overexpression can be considered a contributing factor leading to PE, causing detective uterine spiral artery remodeling and aberrant placental vascularization.
Collapse
Affiliation(s)
- Jingyun Wang
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ping Zhang
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Mengyuan Liu
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
| | - Zhengrui Huang
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xiaofeng Yang
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Yuzhen Ding
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Jia Liu
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China
| | - Shujie Xu
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China
| | - Meiyao He
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Fengxiang Zhang
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China.
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China.
| | - Ruiman Li
- Department of Gynaecology and Obstetrics, The First Affiliate Hospital of Jinan University, Jinan University, No.613 Huangpu Road West, Guangzhou, 510632, China.
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China.
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, No.601 Huangpu Road West, Guangzhou, 510632, China.
| |
Collapse
|
34
|
Gupta S, Petras L, Tufail MU, Rodriguez Salazar JD, Jim B. Hypertension in Pregnancy: What We Now Know. Curr Opin Nephrol Hypertens 2023; 32:153-164. [PMID: 36683540 DOI: 10.1097/mnh.0000000000000857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW Hypertensive disorders of pregnancy remain a highly morbid condition that affects both the mother and fetus, complicate approximately 10% of pregnancies worldwide, and contribute to immediate and long-term cardiovascular outcomes. There is still much to learn regarding pathogenesis and treatment goals. RECENT FINDINGS There is updated information on the pathogenesis of preeclampsia and treatment thresholds for HTN in pregnancy. l-Kynurenine, a metabolite of the essential amino acid l-tryptophan, has been implicated in preeclampsia as decreased levels were found in a uninephrectomized pregnant mouse model of preeclampsia, where replacement of l-kynurenine rescued the preeclamptic state. Further, data from CHIPS (The Control of HTN in Pregnancy Study) and CHAP (Chronic HTN and Pregnancy) trials demonstrate not only the safety of lowering blood pressure to either a diastolic goal of 85 mmHg (CHIPS) or less than 160/105 mmHg (CHAP) without detriment to the fetus but the CHAPS trial has also shown a decrease in the rate of preeclampsia in the treatment group. SUMMARY We will summarize the different types of hypertensive disorders in pregnancy, updates on the pathogenesis of preeclampsia, and appropriate HTN management based on the latest evidence in order to better care for mother and child.
Collapse
Affiliation(s)
- Sonali Gupta
- Division of Nephrology, Department of Medicine, Montefiore Medical Center at Albert Einstein College of Medicine
| | - Lohana Petras
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York, USA
| | - Muhammad Umer Tufail
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York, USA
| | - Juan Diego Rodriguez Salazar
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York, USA
| | - Belinda Jim
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
35
|
Herrock O, Deer E, LaMarca B. Setting a stage: Inflammation during preeclampsia and postpartum. Front Physiol 2023; 14:1130116. [PMID: 36909242 PMCID: PMC9995795 DOI: 10.3389/fphys.2023.1130116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal mortality worldwide. The immune system plays a critical role in normal pregnancy progression; however, inappropriate inflammatory responses have been consistently linked with PE pathophysiology. This inflammatory phenotype consists of activation of the innate immune system, adaptive immune system, and increased inflammatory mediators in circulation. Moreover, recent studies have shown that the inflammatory profile seen in PE persists into the postpartum period. This manuscript aims to highlight recent advances in research relating to inflammation in PE as well as the inflammation that persists postpartum in women after a PE pregnancy. With the advent of the COVID-19 pandemic, there has been an increase in obstetric disorders associated with COVID-19 infection during pregnancy. This manuscript also aims to shed light on the relationship between COVID-19 infection during pregnancy and the increased incidence of PE in these women.
Collapse
Affiliation(s)
- Owen Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
36
|
Xu Q, Johnson KP, Hardiman M, Helmick D, Zeevi A. Loss of anti-AT1R reactivity in ELISA post-adsorption - False reactivity or interference in the assay? Hum Immunol 2023; 84:286-289. [PMID: 36764862 DOI: 10.1016/j.humimm.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/18/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Autoantibodies to Angiotensin II type 1 receptor (AT1R) are associated with detrimental outcomes in organ transplants. However, reports showed that adsorption with latex beads reduced positive anti-AT1R antibodies, suggesting possible false reactivity. To investigate this conundrum, we studied 11 samples positive for AT1R antibodies with an ELISA kit before and after adsorption. Adsorption significantly reduced the measurable level of AT1R antibodies (28.3 ± 9.8 vs. 6.3 ± 3.0 U/ml, p < 0.001). AT1R antibodies were lower when post-adsorption serum was added back at 1:1 ratio to the neat serum compared to the diluent control (8.6 ± 4.2 vs. 18.1 ± 10.3 U/ml, p = 0.02). Sham adsorption with the buffer from Adsorb Out™ kit without beads also suppressed the detection of anti-AT1R antibodies (32.7 ± 9.1 vs. 8.1 ± 3.9 U/ml, p < 0.001). Thus, rather than actively removing nonspecific antibodies by the beads, the adsorption process introduces soluble factors that interfere with the detection of anti-AT1R antibodies with the ELISA kit.
Collapse
Affiliation(s)
- Qingyong Xu
- Department of Pathology, University of Pittsburgh Medical Center, United States.
| | - Kurt P Johnson
- Department of Pathology, University of Pittsburgh Medical Center, United States
| | - Maura Hardiman
- Department of Pathology, University of Pittsburgh Medical Center, United States
| | - Dennis Helmick
- Department of Pathology, University of Pittsburgh Medical Center, United States
| | - Adriana Zeevi
- Department of Pathology, University of Pittsburgh Medical Center, United States
| |
Collapse
|
37
|
AT1 receptor autoantibodies mediate effects of metabolic syndrome on dopaminergic vulnerability. Brain Behav Immun 2023; 108:255-268. [PMID: 36535607 DOI: 10.1016/j.bbi.2022.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The metabolic syndrome has been associated to chronic peripheral inflammation and related with neuroinflammation and neurodegeneration, including Parkinson's disease. However, the responsible mechanisms are unclear. Previous studies have involved the brain renin-angiotensin system in progression of Parkinson's disease and the angiotensin receptor type 1 (AT1) has been recently revealed as a major marker of dopaminergic vulnerability in humans. Dysregulation of tissue renin-angiotensin system is a key common mechanism for all major components of metabolic syndrome. Circulating AT1 agonistic autoantibodies have been observed in several inflammation-related peripheral processes, and activation of AT1 receptors of endothelial cells, dopaminergic neurons and glial cells have been observed to disrupt endothelial blood -brain barrier and induce neurodegeneration, respectively. Using a rat model, we observed that metabolic syndrome induces overactivity of nigral pro-inflammatory renin-angiotensin system axis, leading to increase in oxidative stress and neuroinflammation and enhancing dopaminergic neurodegeneration, which was inhibited by treatment with AT1 receptor blockers (ARBs). In rats, metabolic syndrome induced the increase in circulating levels of LIGHT and other major pro-inflammatory cytokines, and 27-hydroxycholesterol. Furthermore, the rats showed a significant increase in serum levels of proinflammatory AT1 and angiotensin converting enzyme 2 (ACE2) autoantibodies, which correlated with levels of several metabolic syndrome parameters. We also found AT1 and ACE2 autoantibodies in the CSF of these rats. Effects of circulating autoantibodies were confirmed by chronic infusion of AT1 autoantibodies, which induced blood-brain barrier disruption, an increase in the pro-inflammatory renin-angiotensin system activity in the substantia nigra and a significant enhancement in dopaminergic neuron death in two different rat models of Parkinson's disease. Observations in the rat models, were analyzed in a cohort of parkinsonian and non-parkinsonian patients with or without metabolic syndrome. Non-parkinsonian patients with metabolic syndrome showed significantly higher levels of AT1 autoantibodies than non-parkinsonian patients without metabolic syndrome. However, there was no significant difference between parkinsonian patients with metabolic syndrome or without metabolic syndrome, which showed higher levels of AT1 autoantibodies than non-parkinsonian controls. This is consistent with our recent studies, showing significant increase of AT1 and ACE2 autoantibodies in parkinsonian patients, which was related to dopaminergic degeneration and neuroinflammation. Altogether may lead to a vicious circle enhancing the progression of the disease that may be inhibited by strategies against production of these autoantibodies or AT1 receptor blockers (ARBs).
Collapse
|
38
|
Herrock OT, Deer E, Amaral LM, Campbell N, Lemon J, Ingram N, Cornelius DC, Turner TW, Fitzgerald S, Ibrahim T, Dechend R, Wallukat G, LaMarca B. B2 cells contribute to hypertension and natural killer cell activation possibly via AT1-AA in response to placental ischemia. Am J Physiol Renal Physiol 2023; 324:F179-F192. [PMID: 36417275 PMCID: PMC9844978 DOI: 10.1152/ajprenal.00190.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia, new onset hypertension during pregnancy, is associated with activated T helper cells (Th) and B cells secreting agonistic autoantibodies against the angiotensin II type 1 receptor (AT1-AA). The reduced uterine perfusion pressure (RUPP) model of placental ischemia recapitulates these characteristics. We have shown that Th-B cell communication contributes to AT1-AA and symptoms of preeclampsia in the RUPP rat. B2 cells are classical B cells that communicate with Th cells and are then transformed into memory B cells. We hypothesize that B2 cells cause hypertension, natural killer (NK) cell activation, and complement activation during pregnancy through the production of AT1-AA. To test this hypothesis, total splenic B cells and B2 cells were isolated from normal pregnant (NP) or RUPP rats on gestational day (GD)19 and adoptively transferred into GD12 NP rats. A group of recipient rats was treated with a specific inhibitor peptide of AT1-AA. On GD19, mean arterial pressure was measured, tissues were collected, activated NK cells were measured by flow cytometry, and AT1-AA was measured by cardiomyocyte assay. NP recipients of RUPP B cells or RUPP B2 cells had increased mean arterial pressure, AT1-AA, and circulating activated NK cells compared with recipients of NP B cells. Hypertension in NP recipients of RUPP B cells or RUPP B2 was attenuated with AT1-AA blockade. This study demonstrates that B cells and B2 cells from RUPP rats cause hypertension and increased AT1-AA and NK cell activation in response to placental ischemia during pregnancy.NEW & NOTEWORTHY This study demonstrates that placental ischemia-stimulated B2 cells induce hypertension and circulating natural killer cell activation and angiotensin II type 1 receptor production in normal pregnant rats.
Collapse
Affiliation(s)
- Owen T Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - James Lemon
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nicole Ingram
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ty W Turner
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center, HELIOS Clinic, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
39
|
Linge LL, Sugulle M, Wallukat G, Dechend R, Staff AC. Circulating angiotensin II type I receptor - autoantibodies in diabetic pregnancies. J Reprod Immunol 2023; 155:103777. [PMID: 36495655 DOI: 10.1016/j.jri.2022.103777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Pregnant women with either pre-existing or gestational diabetes mellitus are at increased risk of preeclampsia as well as future cardiovascular disease. The renin-angiotensin system is dysregulated in both diabetes mellitus and preeclampsia. In preeclampsia, maternal levels of circulating agonistic autoantibodies against the angiotensin II Type I receptor (AT1-AAs) are increased. Circulating AT1-AAs are thought to contribute to both the pathophysiology of preeclampsia and the increased risk of future cardiovascular disease. Studies exploring AT1-AA in diabetes outside pregnancy suggest their potential for both metabolic and cardiovascular pathogenicity. No studies have investigated AT1-AAs in diabetic pregnancies. We hypothesized elevated maternal circulating AT1-AA levels in pregnancies complicated by any type of diabetes mellitus. Third-trimester maternal serum from 39 women (controls: n = 10; type 1 diabetes: n = 9; type 2 diabetes: n = 10; gestational diabetes=10) were analyzed for AT1-AA using an established bioassay method. Circulating AT1-AAs were present in 70% (7/10) of the controls and 83% (24/29) of the diabetes group (P = 0.399). Presence of AT1-AA was correlated to hsCRP levels (P = 0.036), but neither with maternal circulating angiogenic factors (soluble fms-like tyrosine kinase-1 and placental growth factor), nor with maternal or fetal characteristics indicative of metabolic disease or placental dysfunction. Our study is the first to demonstrate presence of circulating AT1-AAs in pregnant women with any type of diabetes. Our findings suggest AT1-AAs presence in pregnancy independently of placental dysfunction, nuancing the current view on their pathogenicity. Whether AT1-AAs per se contribute to increased risk of adverse pregnancy outcomes and future cardiovascular disease remains currently unanswered.
Collapse
Affiliation(s)
- Lydia Lande Linge
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Gynaecology and Obstetrics, Oslo University Hospital, Oslo, Norway
| | - Meryam Sugulle
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Gynaecology and Obstetrics, Oslo University Hospital, Oslo, Norway.
| | - Gerd Wallukat
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany; Department of Cardiology and Nephrology, HELIOS Klinikum Berlin, Germany
| | - Anne Cathrine Staff
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Gynaecology and Obstetrics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
40
|
Abstract
Primary aldosteronism (PA) is the most common form of secondary hypertension. Although hypertensive disorders seem to affect around 5-10% of pregnancies worldwide, literature counts less than 80 cases of PA diagnosed during the peri-partum period. In this review we discuss about current knowledge on pathophysiology, natural history, diagnosis and treatment of PA in pregnancy. Because of the physiologic changes in the renin-angiotensin-aldosterone system (RAAS) and the contraindication to both confirmatory test and subtype differentiation, diagnosis of PA during pregnancy is challenging and relies mostly on detection of low/suppressed renin and high aldosterone levels. The course of pregnancy in patients with PA is highly variable, ranging from progesterone-induced amelioration of blood pressure (BP) control to severe and resistant hypertension with potential maternal and fetal complications. Mineralcorticoid receptor antagonists (MRA) are the recommended and most effective drugs for treatment of PA. As the anti-androgenic effect of spironolactone can potentially interfere with sexual development, their prescription is not recommended during pregnancy. On the other side, eplerenone, has proven to be safe and effective in 6 pregnant women and may be added to conventional first line drug regimen in presence of resistant hypertension or persistent hypokalemia. Ideally, patients with unilateral forms of PA should undergo adrenalectomy prior to conception, however, when PA is diagnosed during pregnancy and medical therapy fails to adequately control hypertension or its complications, adrenalectomy can be considered during the second trimester in case of unilateral adrenal mass at MRI-scan.
Collapse
Affiliation(s)
- Vittorio Forestiero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Via Genova 3, 10126, Torino, Italy
| | - Elisa Sconfienza
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Via Genova 3, 10126, Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Via Genova 3, 10126, Torino, Italy.
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Via Genova 3, 10126, Torino, Italy
| |
Collapse
|
41
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
42
|
Ashraf UM, Hall DL, Campbell N, Waller JP, Rawls AZ, Solise D, Cockrell K, Bidwell GL, Romero DG, Ojeda NB, LaMarca B, Alexander BT. Inhibition of the AT 1R agonistic autoantibody in a rat model of preeclampsia improves fetal growth in late gestation. Am J Physiol Regul Integr Comp Physiol 2022; 323:R670-R681. [PMID: 36121142 PMCID: PMC9602704 DOI: 10.1152/ajpregu.00122.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/17/2022] [Accepted: 09/09/2022] [Indexed: 02/07/2023]
Abstract
Placenta ischemia, the initiating event in preeclampsia (PE), is associated with fetal growth restriction. Inhibition of the agonistic autoantibody against the angiotensin type 1 receptor AT1-AA, using an epitope-binding inhibitory peptide ('n7AAc') attenuates increased blood pressure at gestational day (G)19 in the clinically relevant reduced uterine perfusion pressure (RUPP) model of PE. Thus we tested the hypothesis that maternal administration of 'n7AAc' does not transfer to the fetus, improves uterine blood flow and fetal growth, and attenuates elevated placental expression of miRNAs implicated in PE and FGR. Sham or RUPP surgery was performed at G14 with vehicle or 'n7AAc' (144 µg/day) administered via an osmotic pump from G14 to G20. Maternal plasma levels of the peptide on G20 were 16.28 ± 4.4 nM, and fetal plasma levels were significantly lower at 1.15 ± 1.7 nM (P = 0.0007). The uterine artery resistance index was significantly elevated in RUPP (P < 0.0001) but was not increased in 'n7AAc'-RUPP or 'n7AAc'-Sham versus Sham. A significant reduction in fetal weight at G20 in RUPP (P = 0.003) was not observed in 'n7AAc'-RUPP. Yet, percent survival was reduced in RUPP (P = 0.0007) and 'n7AAc'-RUPP (P < 0.0002). Correlation analysis indicated the reduction in percent survival during gestation was specific to the RUPP (r = 0.5342, P = 0.043) and independent of 'n7AAc'. Placental miR-155 (P = 0.0091) and miR-181a (P = 0.0384) expression was upregulated in RUPP at G20 but was not elevated in 'n7AAc'-RUPP. Collectively, our results suggest that maternal administration of 'n7AAc' does not alter fetal growth in the RUPP implicating its potential as a therapeutic for the treatment of PE.NEW & NOTEWORTHY The seven amino acid inhibitory peptide to the AT1-AA ('n7AAc') has limited transfer to the fetus at gestational day 20, improves uterine blood flow and fetal growth in the reduced uterine perfusion pressure model of preeclampsia (PE), and does not impair fetal survival during gestation in sham-operated or placental ischemic rats. Collectively, these findings suggest that maternal administration of 'n7AAc' as an effective strategy for the treatment of PE is associated with improved outcomes in the fetus.
Collapse
Affiliation(s)
- Usman M Ashraf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jamarius P Waller
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Adam Z Rawls
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dylan Solise
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kathy Cockrell
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
43
|
Demery-Poulos C, Romero R, Xu Y, Arenas-Hernandez M, Miller D, Tao L, Galaz J, Farias-Jofre M, Bhatti G, Garcia-Flores V, Seyerle M, Tarca AL, Gomez-Lopez N. Pregnancy imparts distinct systemic adaptive immune function. Am J Reprod Immunol 2022; 88:e13606. [PMID: 35989229 PMCID: PMC9648024 DOI: 10.1111/aji.13606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Pregnancy represents a state of systemic immune activation that is primarily driven by alterations in circulating innate immune cells. Recent studies have suggested that cellular adaptive immune components, T cells and B cells, also undergo changes throughout gestation. However, the phenotypes and functions of such adaptive immune cells are poorly understood. Herein, we utilized high-dimensional flow cytometry and functional assays to characterize T-cell and B-cell responses in pregnant and non-pregnant women. METHODS Peripheral blood mononuclear cells from pregnant (n = 20) and non-pregnant (n = 25) women were used for phenotyping of T-cell and B-cell subsets. T-cell proliferation and B-cell activation were assessed by flow cytometry after in vitro stimulation, and lymphocyte cytotoxicity was evaluated by using a cell-based assay. Statistical comparisons were performed with linear mixed-effects models. RESULTS Pregnancy was associated with modestly enhanced basal activation of peripheral CD4+ T cells. Both CD4+ and CD8+ T cells from pregnant women showed increased activation-induced proliferation; yet, a reduced proportion of these cells expressed activation markers compared to non-pregnant women. There were no differences in peripheral lymphocyte cytotoxicity between study groups. A greater proportion of B cells from pregnant women displayed memory-like and activated phenotypes, and such cells exhibited higher activation following stimulation. CONCLUSION Maternal circulating T cells and B cells display distinct responses during pregnancy. The former may reflect the unique capacity of T cells to respond to potential threats without undergoing aberrant activation, thereby preventing systemic inflammatory responses that can lead to adverse perinatal consequences.
Collapse
Affiliation(s)
- Catherine Demery-Poulos
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Centerfor Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Megan Seyerle
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
44
|
Navaneethabalakrishnan S, Smith HL, Arenaz CM, Goodlett BL, McDermott JG, Mitchell BM. Update on Immune Mechanisms in Hypertension. Am J Hypertens 2022; 35:842-851. [PMID: 35704473 PMCID: PMC9527774 DOI: 10.1093/ajh/hpac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/02/2023] Open
Abstract
The contribution of immune cells in the initiation and maintenance of hypertension is undeniable. Several studies have established the association between hypertension, inflammation, and immune cells from the innate and adaptive immune systems. Here, we provide an update to our 2017 American Journal of Hypertension review on the overview of the cellular immune responses involved in hypertension. Further, we discuss the activation of immune cells and their contribution to the pathogenesis of hypertension in different in vivo models. We also highlight existing gaps in the field of hypertension that need attention. The main goal of this review is to provide a knowledge base for translational research to develop therapeutic strategies that can improve cardiovascular health in humans.
Collapse
Affiliation(s)
| | - Hannah L Smith
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Cristina M Arenaz
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Bethany L Goodlett
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Justin G McDermott
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| |
Collapse
|
45
|
Graßhoff H, Fourlakis K, Comdühr S, Riemekasten G. Autoantibodies as Biomarker and Therapeutic Target in Systemic Sclerosis. Biomedicines 2022; 10:2150. [PMID: 36140251 PMCID: PMC9496142 DOI: 10.3390/biomedicines10092150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare connective tissue disorder characterized by immune dysregulation evoking the pathophysiological triad of inflammation, fibrosis and vasculopathy. In SSc, several alterations in the B-cell compartment have been described, leading to polyclonal B-cell hyperreactivity, hypergammaglobulinemia and autoantibody production. Autoreactive B cells and autoantibodies promote and maintain pathologic mechanisms. In addition, autoantibodies in SSc are important biomarkers for predicting clinical phenotype and disease progression. Autoreactive B cells and autoantibodies represent potentially promising targets for therapeutic approaches including B-cell-targeting therapies, as well as strategies for unselective and selective removal of autoantibodies. In this review, we present mechanisms of the innate immune system leading to the generation of autoantibodies, alterations of the B-cell compartment in SSc, autoantibodies as biomarkers and autoantibody-mediated pathologies in SSc as well as potential therapeutic approaches to target these.
Collapse
Affiliation(s)
- Hanna Graßhoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany
| | | | | | | |
Collapse
|
46
|
Tang WW, Naga Prasad SV. Autoantibodies and Cardiomyopathy: Focus on Beta-1 Adrenergic Receptor Autoantibodies. J Cardiovasc Pharmacol 2022; 80:354-363. [PMID: 35323150 PMCID: PMC9452444 DOI: 10.1097/fjc.0000000000001264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
ABSTRACT Antibody response to self-antigens leads to autoimmune response that plays a determinant role in cardiovascular disease outcomes including dilated cardiomyopathy (DCM). Although the origins of the self-reactive endogenous autoantibodies are not well-characterized, it is believed to be triggered by tissue injury or dysregulated humoral response. Autoantibodies that recognize G protein-coupled receptors are considered consequential because they act as modulators of downstream receptor signaling displaying a wide range of unique pharmacological properties. These wide range of pharmacological properties exhibited by autoantibodies has cellular consequences that is associated with progression of disease including DCM. Increase in autoantibodies recognizing beta-1 adrenergic receptor (β1AR), a G protein-coupled receptor critical for cardiac function, is observed in patients with DCM. Cellular and animal model studies have indicated pathological roles for the β1AR autoantibodies but less is understood about the molecular basis of their modulatory effects. Despite the recognition that β1AR autoantibodies could mediate deleterious outcomes, emerging evidence suggests that not all β1AR autoantibodies are deleterious. Recent clinical studies show that β1AR autoantibodies belonging to the IgG3 subclass is associated with beneficial cardiac outcomes in patients. This suggests that our understanding on the roles the β1AR autoantibodies play in mediating outcomes is not well-understood. Technological advances including structural determinants of antibody binding could provide insights on the modulatory capabilities of β1AR autoantibodies in turn, reflecting their diversity in mediating β1AR signaling response. In this study, we discuss the significance of the diversity in signaling and its implications in pathology.
Collapse
Affiliation(s)
- W.H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH
| | - Sathyamangla V. Naga Prasad
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
47
|
Singh KD, Karnik SS. Structural perspectives on the mechanism of signal activation, ligand selectivity and allosteric modulation in angiotensin receptors: IUPHAR Review 34. Br J Pharmacol 2022; 179:4461-4472. [PMID: 35318654 PMCID: PMC9398925 DOI: 10.1111/bph.15840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
Functional advances have guided our knowledge of physiological and fatal pathological mechanisms of the hormone angiotensin II (AngII) and its antagonists. Such studies revealed that tissue response to a given dose of the hormone or its antagonist depends on receptors that engage the ligand. Thus, we need to know much more about the structures of receptor-ligand complexes at high resolution. Recently, X-ray structures of both AngII receptors (AT1 and AT2 receptors) bound to peptide and non-peptide ligands have been elucidated, providing new opportunities to examine the dynamic fluxes in the 3D architecture of the receptors, as the basis of ligand selectivity, efficacy, and regulation of the molecular functions of the receptors. Constituent structural motifs cooperatively transform ligand selectivity into specific functions, thus conceptualizing the primacy of the 3D structure over individual motifs of receptors. This review covers the new data elucidating the structural dynamics of AngII receptors and how structural knowledge can be transformative in understanding the mechanisms underlying the physiology of AngII.
Collapse
Affiliation(s)
- Khuraijam Dhanachandra Singh
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Sadashiva S. Karnik
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
48
|
Tossetta G, Fantone S, delli Muti N, Balercia G, Ciavattini A, Giannubilo SR, Marzioni D. Preeclampsia and severe acute respiratory syndrome coronavirus 2 infection: a systematic review. J Hypertens 2022; 40:1629-1638. [PMID: 35943095 PMCID: PMC10860893 DOI: 10.1097/hjh.0000000000003213] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the coronavirus disease 2019 (COVID-19) disease that has rapidly spread worldwide, causing hundreds of thousand deaths. Normal placentation is characterized by many processes strictly regulated during pregnancy. If placentation is impaired, it can lead to gestational disorders, such as preeclampsia that is a multisystem disorder that occurs in 2-8% of pregnancies worldwide. METHODS We performed a systematic search to understand the potential involvement of SARS-CoV-2 in preeclampsia onset using the databases, PubMed and Web of Science until 31 January 2022. RESULTS SARS-CoV-2 infection not only causes damage to the respiratory system but also can infect human placenta cells impairing pivotal processes necessary for normal placenta development. The inflammatory response trigged by COVID-19 disease is very similar to that one found in preeclampsia pregnancies suggesting a possible link between SARS-CoV-2 infection and preeclampsia onset during pregnancy. CONCLUSION Some studies showed that pregnancies affected by COVID-19 had higher incidence of preeclampsia compared with SARS-CoV-2-negative ones. However, increased blood pressure found in COVID-19 pregnancies does not allow to associate COVID-19 to preeclampsia as hypertension is a common factor to both conditions. At present, no diagnostic tools are available to discriminate real preeclampsia from preeclampsia-like syndrome in patients with SARS-CoV-2 infection. Thus, new specific diagnostic tools are necessary to assure an appropriate diagnosis of preeclampsia in these patients, especially in case of severe COVID-19 disease.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Umberto I Hospital
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Umberto I Hospital
| | - Nicola delli Muti
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Umberto I Hospital, Ancona, Italy
| | - Giancarlo Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Umberto I Hospital, Ancona, Italy
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Umberto I Hospital
| |
Collapse
|
49
|
Trakarnvanich T, Ngamvichchukorn T, Susantitaphong P. Incidence of acute kidney injury during pregnancy and its prognostic value for adverse clinical outcomes: A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e29563. [PMID: 35905231 PMCID: PMC9333530 DOI: 10.1097/md.0000000000029563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) that develops during pregnancy results from pregnancy-induced hypertension, hemorrhage, and sepsis, associated with morbidity and mortality in the fetus and mother. This meta-analysis was conducted to evaluate the incidence of pregnancy-related AKI (PR-AKI) and adverse clinical outcomes. METHODS PubMed and Scopus were systematically searched for studies published between 1980 and 2021. We included cross-sectional, retrospective, and prospective cohort studies that reported the incidence of PR-AKI as well as adverse fetal and maternal clinical outcomes. A random-effects model meta-analysis was performed to generate summary estimates. RESULTS The meta-analysis included 31 studies (57,529,841 participants). The pooled incidence of PR-AKI was 2.0% (95% confidence interval [CI] 1.0-3.7). Only 49.3% of patients received antenatal care. The most common cause of PR-AKI was preeclampsia (36.6%, 95% CI 29.1-44.7). The proportion of patients requiring hemodialysis was 37.2% (95% CI 26.0-49.9). More than 70% of patients had complete recovery of renal function, while 8.5% (95% CI 4.7-14.8) remained dependent on dialysis. The pooled mortality rate of PR-AKI was 12.7% (95% CI 9.0-17.7). In addition, fetal outcomes were favorable, with an alive birth rate of 70.0% (95% CI 61.2-77.4). However, the rate of abortion and/or stillbirth was approximately 25.4% (95% CI 18.1-34.4), and the rate of intrauterine death was 18.6% (95% CI 12.8-26.2). CONCLUSIONS Although the incidence of PR-AKI is not high, this condition has a high impact on morbidity and mortality in both fetal and maternal outcomes. Early prevention and treatment from health care professionals are needed in PR-AKI, especially in the form of antenatal care and preeclampsia medication.
Collapse
Affiliation(s)
- Thananda Trakarnvanich
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Tanun Ngamvichchukorn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Paweena Susantitaphong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Research Unit for Metabolic Bone Disease in CKD patients, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Paweena Susantitaphong, 1873 Rama IV Road, Pathumwan, Bangkok, Thailand (e-mail: )
| |
Collapse
|
50
|
Barr LC, Liblik K, Johri AM, Smith GN. Maternal Cardiovascular Function Following a Pregnancy Complicated by Preeclampsia. Am J Perinatol 2022; 39:1055-1064. [PMID: 33321533 DOI: 10.1055/s-0040-1721694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preeclampsia is a hypertensive pregnancy complication with an unknown etiology and high maternal burden worldwide. Burgeoning research has linked preeclampsia to adverse maternal health outcomes remote from pregnancy; however, the intermediary mechanisms responsible for this association have not been sufficiently established. In the present narrative review, we summarize leading evidence of structural and functional cardiovascular changes associated with prior preeclampsia, and how these changes may be linked to future maternal disease. KEY POINTS: · Prior preeclampsia is associated with subclinical structural and functional vascular changes remote from pregnancy.. · Maternal cardiac adaptations to preeclampsia may have long-term implications on cardiovascular health.. · Clinicians have an opportunity to minimize maternal disease risk following preeclampsia..
Collapse
Affiliation(s)
- Logan C Barr
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Kiera Liblik
- Cardiovascular Imaging Network at Queen's, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Amer M Johri
- Cardiovascular Imaging Network at Queen's, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Graeme N Smith
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|