1
|
Eslahi H, Saravani M, Shahraki M, Payandeh A, Rezaei M, Ghasemi M, Salimi S, Sargazi-Taghazi M. Investigation of The Placental Levels of Vascular Endothelial Growth Factor B and Selenoprotein P and Their Relation with Birth Weight in Patients with Pre-eclampsia: A Case-Control Study. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2025; 19:246-250. [PMID: 40200785 PMCID: PMC11976889 DOI: 10.22074/ijfs.2024.2030292.1698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/09/2024] [Accepted: 11/20/2024] [Indexed: 04/10/2025]
Abstract
BACKGROUND Pre-eclampsia is a serious medical condition characterised by high blood pressure and signs of organ damage that is associated with adverse pregnancy outcomes, including low birth weight. Selenoprotein P (SELENOP) and vascular endothelial growth factor B (VEGF-B) are antioxidants that can improve the condition of this disease. This study aims to investigate the placental levels of SELENOP and VEGF-B, and their association with birth weight in pregnant women with pre-eclampsia compared to healthy pregnant women. MATERIALS AND METHODS This case-control study enrolled 30 pregnant women with pre-eclampsia as the case group and 30 healthy pregnant women as the control group. Demographic information and anthropometric indices were collected and recorded in forms. Placental levels of SELENOP and VEGF-B were assessed by a commercial human kit and based on enzyme-linked immunosorbent assay (ELISA). P<0.05 indicated statistical significance. RESULTS The mean placental level of VEGF-B in pregnant women with pre-eclampsia was lower than the healthy group (P=0.001) as was the mean placental level of SELENOP compared to the healthy group (P=0.048). No significant correlation existed between placental levels of SELENOP (r=0.253, P=0.051) and VEGF-B (r=0.671, P=0.056) with birth weight. CONCLUSION Our findings showed that pregnant women with pre-eclampsia had lower levels of VEGF-B and SELENOP compared to healthy pregnant women. The findings may assist with pre-eclampsia diagnosis, management, and prediction, and benefit mothers and babies.
Collapse
Affiliation(s)
- Hadi Eslahi
- Children and Adolescents Health Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohsen Saravani
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Genetics of Non-communicable Disease Research Center, Zahedan of UniversityMedical Sciences, Zahedan, Iran
| | - Mansour Shahraki
- Children and Adolescents Health Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Abolfazl Payandeh
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahnaz Rezaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Genetics of Non-communicable Disease Research Center, Zahedan of UniversityMedical Sciences, Zahedan, Iran
| | - Marzieh Ghasemi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeedeh Salimi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahjob Sargazi-Taghazi
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
2
|
Jones K, Smith S, Smith J, Castillo A, Burkes A, Howard A, Garvin MM, Bolton JL, Colon-Perez L, Cunningham MW. Postpartum dams exposed to a low-resource environment display neuroinflammation, elevated corticosterone, and anhedonia-like behavior. J Appl Physiol (1985) 2025; 138:666-680. [PMID: 39884662 DOI: 10.1152/japplphysiol.00871.2024] [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: 11/07/2024] [Revised: 11/26/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Women living in an impoverished environment after birth have an increased risk of developing postpartum depression (PP-Dep) and hypertension (PP-HTN). The mechanisms underlying these heightened risks are unknown and understudied. To examine the relation between reduced environmental resources, PP-Dep, and PP-HTN, postpartum rodent dams were exposed to the low-resource limited bedding and nesting (LBN) chronic stress model during weaning. Postpartum dams were divided into control (CTL) and experimental (LBN) groups, in which the experimental group experienced LBN. At 6 wks postpartum, blood pressure, sucrose preference tests (a proxy for anhedonia and depression), corticosterone, and markers of neuroinflammation were measured. We hypothesized that postpartum dams exposed to LBN will have increased corticosterone, neuroinflammation, depression-like behaviors, and HTN. Results show that postpartum dams exposed to an impoverished environment exhibit decreased sucrose preference, increased circulating corticosterone, and elevated neuroinflammation (∼150% increased TNF-α and astrocyte activation in the cerebrum). No changes in blood pressure were observed. However, there was a strong correlation between postpartum blood pressure and corticosterone and blood pressure and TNF-α levels. Importantly, this study provides insights into the pathology and development of PP-HTN and PP-Dep in the postpartum period, which will enable the discovery of novel therapeutic approaches.NEW & NOTEWORTHY Postpartum dams exposed to a low-resource environment experience anhedonia, elevated corticosterone, and neuroinflammation. Increases in corticosterone and neuroinflammation may contribute to the development of postpartum depression (PP-Dep) and postpartum hypertension (PP-HTN). Healthcare providers should consider asking questions about the social economic status and accessibility of resources for women after pregnancy. This study advocates for extended postpartum care beyond traditional care and better implementation of assessments for PP-Dep and PP-HTN.
Collapse
Affiliation(s)
- Kylie Jones
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Savanna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Jonna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Angie Castillo
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Allison Burkes
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Ahfiya Howard
- School of Social Work, East Texas A&M University, Texas, United States
| | - Madison M Garvin
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States
| | - Luis Colon-Perez
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Mark W Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
3
|
Deer E, Herrock O, Simmons K, Campbell N, Amaral L, Zheng B, Morris R, Wallace K, Cleveland ELH, Belk S, Dodd C, LaMarca B. Hypertension and Cognitive Dysfunction in a Pregnant Rat Model of PE; a Role for CD4+ T Cells. Am J Reprod Immunol 2024; 92:e13935. [PMID: 39427297 DOI: 10.1111/aji.13935] [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: 05/10/2024] [Revised: 08/27/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE Preeclampsia (PE) is associated with hypertension (HTN) during pregnancy and activated CD4+ T cells, inflammatory cytokines, and autoantibodies to the angiotensin II type I receptor (AT1-AA). Having had COVID-19 (CV) during pregnancy is associated with an increased incidence of a PE-like phenotype. Both PE and CV have long-lasting neurological implications and studies show that nonpregnant COVID patients produce AT1-AA. We have shown that CD4+ T cells from PE women cause a PE phenotype in nude athymic rats. In this study, we sought to examine the role of CD4+ T cells from PE with a CV History (Hx) to contribute to a PE phenotype and to determine the importance of CD4+ T cells in cognitive dysfunction during pregnancy. METHODS At delivery, blood and placentas were collected, and one million placental CD4+ T cells from each PE and each normotensive patient, with (NT) or without (NP) a CV (Hx) during pregnancy, were isolated, purified, and injected i.p. into a gestational day (GD) 12 pregnant nude athymic rat (one patient/rat). At GD19, blood pressure (MAP) and circulating factors were assessed in recipient rats. Cognitive function and memory were assessed using Novel Object Recognition and Barnes Maze tests, respectively. Placental ACE-2 activity and AT1-AA were measured from COVID Hx patients. A one- or two-way ANOVA with Bonferroni's multiple comparisons test was used for statistical analysis. RESULTS Blood pressure was increased in patients with PE, with or without COVID, compared to NT patients. There were no significant changes in placental ACE activity in patients with COVID Hx with or without PE. AT1-AA was elevated in PE patients and in both PE and NT COVID Hx compared to control NP. In pregnant recipient rats, MAP increased in CV Hx PE (113 ± 2, n = 8) compared to CV Hx NT (101 ± 5, n = 6). PE and PE CV Hx CD4+ T Cell recipient rats exhibited impaired memory and cognitive dysfunction (p < 0.05), compared to control groups. Recipient rats of PE CV Hx CD4+ T cells had elevated AT1-AA compared to NT CV Hx recipients. Both COVID Hx groups and recipients of PE CD4+ T cells had elevated TNF alpha compared to NP. CONCLUSION Our findings indicate that pregnant patients with a Hx of COVID during pregnancy produce AT1-AA, with or without PE. Recipients of CD4+ T cells from PE with or without a CV Hx during pregnancy cause HTN and elevated AT1-AA. TNF-α is elevated in PE and in CV Hx NT and PE recipients. Interestingly, recipients of T cells from PE patients with or without a Hx of CV had worse cognitive function during pregnancy, compared to recipient rats of NP CD4+ T cells. These data demonstrate the importance of CD4+ T cells in HTN and impaired neurological function during PE in the presence or absence of a prior COVID-19 infection during pregnancy.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kimberly Simmons
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lorena Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Baoying Zheng
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rachael Morris
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Sheila Belk
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Cameronne Dodd
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
4
|
Elgazzaz M, Woodham PC, Maher J, Faulkner JL. Implications of pregnancy on cardiometabolic disease risk: preeclampsia and gestational diabetes. Am J Physiol Cell Physiol 2024; 327:C646-C660. [PMID: 39010840 PMCID: PMC11427017 DOI: 10.1152/ajpcell.00293.2024] [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: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Cardiometabolic disorders, such as obesity, insulin resistance, and hypertension, prior to and within pregnancy are increasing in prevalence worldwide. Pregnancy-associated cardiometabolic disease poses a great risk to the short- and long-term well-being of the mother and offspring. Hypertensive pregnancy, notably preeclampsia, as well as gestational diabetes are the major diseases of pregnancy growing in prevalence as a result of growing cardiometabolic disease prevalence. The mechanisms whereby obesity, diabetes, and other comorbidities lead to preeclampsia and gestational diabetes are incompletely understood and continually evolving in the literature. In addition, novel therapeutic avenues are currently being explored in these patients to offset cardiometabolic-induced adverse pregnancy outcomes in preeclamptic and gestational diabetes pregnancies. In this review, we discuss the emerging pathophysiological mechanisms of preeclampsia and gestational diabetes in the context of cardiometabolic risk as well as the most recent preclinical and clinical updates in the pathogenesis and treatment of these conditions.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Padmashree C Woodham
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - James Maher
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
5
|
Al-Nuaimi AMA. Role of hematological indices in predicting preeclampsia and its severity: retrospective case-control study. Medicine (Baltimore) 2024; 103:e38557. [PMID: 38905404 PMCID: PMC11192010 DOI: 10.1097/md.0000000000038557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/23/2024] Open
Abstract
Preeclampsia (PE) is a serious condition that threatens pregnancy with severe sequelae on both the mother and infant. Early detection of PE will lead to favorable outcomes, and using readily available markers like hematological indices is an attractive choice. Examine the diagnostic utility of hematological indices in pregnant women to predict preeclampsia and its severity. In a retrospective case-control study that included 252 women, all had their complete blood picture evaluated during their first and third trimesters as part of their outpatient antenatal care during their pregnancy. They were also divided into 3 groups: healthy pregnant women (control), non-severe PE, and severe PE, each involving 84 women. The changes in platelet to lymphocyte ratio (PLR) between 1st and 3rd trimesters showed an excellent ability to differentiate between severe PE and control (area under the curve = 0.954, cutoff ≤ -5.45%) and a good ability to differentiate between severe PE and non-severe PE (area under the curve = 0.841, cutoff ≤ -7.89%). Neutrophil to lymphocyte ratio showed a good to excellent ability to differentiate between severe PE and non-severe PE compared to control in the first and third trimesters and the percentage change between them. Changes in neutrophil to lymphocyte ratio and PLR strongly predict preeclampsia and its severity since they offer more predictive values than measuring NLP and PLR at different stages of pregnancy individually.
Collapse
|
6
|
Svigkou A, Katsi V, Kordalis VG, Tsioufis K. The Molecular Basis of the Augmented Cardiovascular Risk in Offspring of Mothers with Hypertensive Disorders of Pregnancy. Int J Mol Sci 2024; 25:5455. [PMID: 38791492 PMCID: PMC11121482 DOI: 10.3390/ijms25105455] [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] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The review examines the impact of maternal preeclampsia (PE) on the cardiometabolic and cardiovascular health of offspring. PE, a hypertensive disorder of pregnancy, is responsible for 2 to 8% of pregnancy-related complications. It significantly contributes to adverse outcomes for their infants, affecting the time of birth, the birth weight, and cardiometabolic risk factors such as blood pressure, body mass index (BMI), abdominal obesity, lipid profiles, glucose, and insulin. Exposure to PE in utero predisposes offspring to an increased risk of cardiometabolic diseases (CMD) and cardiovascular diseases (CVD) through mechanisms that are not fully understood. The incidence of CMD and CVD is constantly increasing, whereas CVD is the main cause of morbidity and mortality globally. A complex interplay of genes, environment, and developmental programming is a plausible explanation for the development of endothelial dysfunction, which leads to atherosclerosis and CVD. The underlying molecular mechanisms are angiogenic imbalance, inflammation, alterations in the renin-angiotensin-aldosterone system (RAAS), endothelium-derived components, serotonin dysregulation, oxidative stress, and activation of both the hypothalamic-pituitary-adrenal axis and hypothalamic-pituitary-gonadal axis. Moreover, the potential role of epigenetic factors, such as DNA methylation and microRNAs as mediators of these effects is emphasized, suggesting avenues for future research and therapeutic interventions.
Collapse
Affiliation(s)
| | - Vasiliki Katsi
- Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece;
| | - Vasilios G. Kordalis
- School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Konstantinos Tsioufis
- Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece;
| |
Collapse
|
7
|
Couture C, Brien ME, Rechtzigel J, Ling S, Ledezma-Soto C, Duran Bishop G, Boufaied I, Dal Soglio D, Rey E, McGraw S, Graham CH, Girard S. Predictive biomarkers and initial analysis of maternal immune alterations in postpartum preeclampsia reveal an immune-driven pathology. Front Immunol 2024; 15:1380629. [PMID: 38745664 PMCID: PMC11091301 DOI: 10.3389/fimmu.2024.1380629] [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: 02/01/2024] [Accepted: 03/13/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Postpartum preeclampsia (PPPE) is an under-diagnosed condition, developing within 48 hours to 6 weeks following an uncomplicated pregnancy. The etiology of PPPE is still unknown, leaving patients vulnerable and making the identification and treatment of patients requiring postpartum care an unmet need. We aimed to understand the immune contribution to PPPE at the time of diagnosis, as well as uncover the predictive potential of perinatal biomarkers for the early postnatal identification of high-risk patients. Methods Placentas were collected at delivery from uncomplicated pregnancies (CTL) and PPPE patients for immunohistochemistry analysis. In this initial study, blood samples in PPPE patients were collected at the time of PPPE diagnosis (48h-25 days postpartum; mean 7.4 days) and compared to CTL blood samples taken 24h after delivery. Single-cell transcriptomics, flow cytometry, intracellular cytokine staining, and the circulating levels of inflammatory mediators were evaluated in the blood. Results Placental CD163+ cells and 1st trimester blood pressures can be valuable non-invasive and predictive biomarkers of PPPE with strong clinical application prospects. Furthermore, changes in immune cell populations, as well as cytokine production by CD14+, CD4+, and CD8+ cells, suggested a dampened response with an exhausted phenotype including decreased IL1β, IL12, and IFNγ as well as elevated IL10. Discussion Understanding maternal immune changes at the time of diagnosis and prenatally within the placenta in our sizable cohort will serve as groundwork for pre-clinical and clinical research, as well as guiding clinical practice for example in the development of immune-targeted therapies, and early postnatal identification of patients who would benefit from more thorough follow-ups and risk education in the weeks following an uncomplicated pregnancy.
Collapse
Affiliation(s)
- Camille Couture
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Jade Rechtzigel
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - SuYun Ling
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Cecilia Ledezma-Soto
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Ines Boufaied
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Dorothée Dal Soglio
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, QC, Canada
| | - Evelyne Rey
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| | - Serge McGraw
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
8
|
Griffin A, Bowles T, Solis L, Railey T, Beauti S, Robinson R, Spencer SK, Shaffery JP, Wallace K. Maternal immune suppression during pregnancy does not prevent abnormal behavior in offspring. Biol Sex Differ 2024; 15:27. [PMID: 38532505 DOI: 10.1186/s13293-024-00600-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Offspring of hypertensive disorders of pregnancy are at an increased risk of developing neurodevelopmental and neurobehavioral disorders compared to offspring from non-affected pregnancies. Using rodent models of Preeclampsia (PreE; new onset of hypertension after 20 weeks gestation) and HELLP (hemolysis, elevated liver enzymes, and low platelets), we studied the behavioral outcome of their offspring in adolescence. METHODS A subset of dams received Orencia, a T-cell activation inhibitor, as T cells have been associated with the induction of hypertension and inflammation during pregnancy. We hypothesized that offspring from hypertensive dams would experience adverse behavioral outcomes in social, cognitive, locomotor, and anxiety tests, and offspring from dams treated with Orencia would demonstrate less adverse behaviors. RESULTS Male offspring of PreE + Orencia dams (p < 0.05) and female offspring from HELLP + Orencia dams (p < 0.05) spent more time playing compared to normal pregnant offspring. All offspring from hypertensive and Orencia-treated dams performed worse on the Barnes Maze test compared to normal pregnant. We also measured adult (postnatal day > 60) myelin basic protein (MBP) and NeuN expression in both the prefrontal cortex and hippocampus. In the hippocampus and prefrontal cortex, there was no difference in expression of either MBP or NeuN in all groups regardless of sex. CONCLUSION The results from this study suggest that offspring of hypertensive disorders of pregnancy have behavioral changes, specifically cognitive differences. This study has shown that there is a sex dependent difference in offspring neurobehavioral development, influenced in part by the type of hypertensive disorder of pregnancy, and alterations in the maternal immune system.
Collapse
Affiliation(s)
- Ashley Griffin
- Program in Neuroscience, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Teylor Bowles
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Lucia Solis
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Teryn Railey
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Samer Beauti
- Master's in Biomedical Science program, School of Graduate Studies in Health Sciences, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Reanna Robinson
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Shauna-Kay Spencer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - James P Shaffery
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
- Department of Obstetrics & Gynecology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
9
|
Wu Q, Ying X, Yu W, Li H, Wei W, Lin X, Yang M, Zhang X. Comparison of immune-related gene signatures and immune infiltration features in early- and late-onset preeclampsia. J Gene Med 2024; 26:e3676. [PMID: 38362844 DOI: 10.1002/jgm.3676] [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: 11/23/2023] [Revised: 01/02/2024] [Accepted: 01/28/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Preeclampsia, a severe pregnancy syndrome, is widely accepted divided into early- and late-onset preeclampsia (EOPE and LOPE) based on the onset time of preeclampsia, with distinct pathophysiological origins. However, the molecular mechanism especially immune-related mechanisms for EOPE and LOPE is currently obscure. In the present study, we focused on placental immune alterations between EOPE and LOPE and search for immune-related biomarkers that could potentially serve as potential therapeutic targets through bioinformatic analysis. METHODS The gene expression profiling data was obtained from the Gene Expression Omnibus database. ESTIMATE algorithm and Gene Set Enrichment Analysis were employed to evaluate the immune status. The intersection of differentially expressed genes in GSE74341 series and immune-related genes set screened differentially expressed immune-related genes. Protein-protein interaction network and random forest were used to identify hub genes with a validation by a quantitative real-time PCR. Kyoto Encyclopedia of Genes and Genomes pathways, Gene Ontology and gene set variation analysis were utilized to conduct biological function and pathway enrichment analyses. Single-sample gene set enrichment analysis and CIBERSORTx tools were employed to calculate the immune cell infiltration score. Correlation analyses were evaluated by Pearson correlation analysis. Hub genes-miRNA network was performed by the NetworkAnalyst online tool. RESULTS Immune score and stromal score were all lower in EOPE samples. The immune system-related gene set was significantly downregulated in EOPE compared to LOPE samples. Four hub differentially expressed immune-related genes (IL15, GZMB, IL1B and CXCL12) were identified based on a protein-protein interaction network and random forest. Quantitative real-time polymerase chain reaction validated the lower expression levels of four hub genes in EOPE compared to LOPE samples. Immune cell infiltration analysis found that innate and adaptive immune cells were apparent lacking in EOPE samples compared to LOPE samples. Cytokine-cytokine receptor, para-inflammation, major histocompatibility complex class I and T cell co-stimulation pathways were significantly deficient and highly correlated with hub genes. We constructed a hub genes-miRNA regulatory network, revealing the correlation between hub genes and hsa-miR-374a-5p, hsa-miR-203a-3p, hsa-miR-128-3p, hsa-miR-155-3p, hsa-miR-129-2-3p and hsa-miR-7-5p. CONCLUSIONS The innate and adaptive immune systems were severely impaired in placentas of EOPE. Four immune-related genes (IL15, GZMB, IL1B and CXCL12) were closely correlated with immune-related pathogenesis of EOPE. The result of our study may provide a new basis for discriminating between EOPE and LOPE and acknowledging the role of the immune landscape in the eventual interference and tailored treatment of EOPE.
Collapse
Affiliation(s)
- Quanfeng Wu
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| | - Xiang Ying
- Department of Gynecology and Obstetrics, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Huanxi Li
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Wei
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueyan Lin
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Meilin Yang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| |
Collapse
|
10
|
Rengarajan A, Austin JL, Stanic AK, Patankar MS, Boeldt DS. Mononuclear Cells Negatively Regulate Endothelial Ca 2+ Signaling. Reprod Sci 2023; 30:2292-2301. [PMID: 36717462 DOI: 10.1007/s43032-023-01164-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/24/2022] [Indexed: 02/01/2023]
Abstract
Endothelial Ca2+ signaling has important roles to play in maintaining pregnancy associated vasodilation in the utero-placenta. Inflammatory cytokines, often elevated in vascular complications of pregnancy, negatively regulate ATP-stimulated endothelial Ca2+ signaling and associated nitric oxide production. However, the role of direct engagement of immune cells on endothelial Ca2+ signaling and therefore endothelial function is unclear. To model immune-endothelial interactions, herein, we evaluate the effects of peripheral blood mononuclear cells (PBMCs) in short-term interaction with human umbilical vein endothelial cells (HUVECs) on agonist-stimulated Ca2+ signaling in HUVECs. We find that mononuclear cells (10:1 and 25:1 mononuclear: HUVEC) cause decreased ATP-stimulated Ca2+ signaling; worsened by activated mononuclear cells possibly due to increased cytokine secretion. Additionally, monocytes, natural killers, and T-cells cause decrease in ATP-stimulated Ca2+ signaling using THP-1 (monocyte), NKL (natural killer cells), and Jurkat (T-cell) cell lines, respectively. PBMCs with Golgi-restricted protein transport prior to interaction with endothelial cells display rescue in Ca2+ signaling, strongly suggesting that secreted proteins from PBMCs mediate changes in HUVEC Ca2+ signaling. We propose that endothelial cells from normal pregnancy interacting with PBMCs may model preeclamptic endothelial-immune interaction and resultant endothelial dysfunction.
Collapse
Affiliation(s)
- Aishwarya Rengarajan
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, Perinatal Research Laboratories, 7E UnityPoint Health-Meriter Hospital, 202 South Park St, Madison, WI, 53715, USA
| | - Jason L Austin
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, Perinatal Research Laboratories, 7E UnityPoint Health-Meriter Hospital, 202 South Park St, Madison, WI, 53715, USA
| | - Aleksandar K Stanic
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, Perinatal Research Laboratories, 7E UnityPoint Health-Meriter Hospital, 202 South Park St, Madison, WI, 53715, USA
| | - Manish S Patankar
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, Perinatal Research Laboratories, 7E UnityPoint Health-Meriter Hospital, 202 South Park St, Madison, WI, 53715, USA
| | - Derek S Boeldt
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, Perinatal Research Laboratories, 7E UnityPoint Health-Meriter Hospital, 202 South Park St, Madison, WI, 53715, USA.
| |
Collapse
|
11
|
Bai L, Guo Y, Gong J, Li Y, Huang H, Meng Y, Liu X. Machine learning and bioinformatics framework integration reveal potential characteristic genes related to immune cell infiltration in preeclampsia. Front Physiol 2023; 14:1078166. [PMID: 37389124 PMCID: PMC10300062 DOI: 10.3389/fphys.2023.1078166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction: Preeclampsia is a disease that affects both the mother and child, with serious consequences. Screening the characteristic genes of preeclampsia and studying the placental immune microenvironment are expected to explore specific methods for the treatment of preeclampsia and gain an in-depth understanding of the pathological mechanism of preeclampsia. Methods: We screened for differential genes in preeclampsia by using limma package. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, disease ontology enrichment, and gene set enrichment analyses were performed. Analysis and identification of preeclampsia biomarkers were performed by using the least absolute shrinkage and selection operator regression model, support vector machine recursive feature elimination, and random forest algorithm. The CIBERSORT algorithm was used to analyze immune cell infiltration. The characteristic genes were verified by RT-qPCR. Results: We identified 73 differential genes, which mainly involved in reproductive structure and system development, hormone transport, etc. KEGG analysis revealed emphasis on cytokine-cytokine receptor interactions and interleukin-17 signaling pathways. Differentially expressed genes were dominantly concentrated in endocrine system diseases and reproductive system diseases. Our findings suggest that LEP, SASH1, RAB6C, and FLT1 can be used as placental markers for preeclampsia and they are associated with various immune cells. Conclusion: The differentially expressed genes in preeclampsia are related to inflammatory response and other pathways. Characteristic genes, LEP, SASH1, RAB6C, and FLT1 can be used as diagnostic and therapeutic targets for preeclampsia, and they are associated with immune cell infiltration. Our findings contribute to the pathophysiological mechanism exploration of preeclampsia. In the future, the sample size needs to be expanded for data analysis and validation, and the immune cells need to be further validated.
Collapse
Affiliation(s)
- Lilian Bai
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyan Guo
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junxing Gong
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yuchen Li
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hefeng Huang
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicong Meng
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinmei Liu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Liang TY, Lu LH, Tang SY, Zheng ZH, Shi K, Liu JQ. Current status and prospects of basic research and clinical application of mesenchymal stem cells in acute respiratory distress syndrome. World J Stem Cells 2023; 15:150-164. [PMID: 37180997 PMCID: PMC10173811 DOI: 10.4252/wjsc.v15.i4.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and clinically devastating disease that causes respiratory failure. Morbidity and mortality of patients in intensive care units are stubbornly high, and various complications severely affect the quality of life of survivors. The pathophysiology of ARDS includes increased alveolar-capillary membrane permeability, an influx of protein-rich pulmonary edema fluid, and surfactant dysfunction leading to severe hypoxemia. At present, the main treatment for ARDS is mechanical treatment combined with diuretics to reduce pulmonary edema, which primarily improves symptoms, but the prognosis of patients with ARDS is still very poor. Mesenchymal stem cells (MSCs) are stromal cells that possess the capacity to self-renew and also exhibit multilineage differentiation. MSCs can be isolated from a variety of tissues, such as the umbilical cord, endometrial polyps, menstrual blood, bone marrow, and adipose tissues. Studies have confirmed the critical healing and immunomodulatory properties of MSCs in the treatment of a variety of diseases. Recently, the potential of stem cells in treating ARDS has been explored via basic research and clinical trials. The efficacy of MSCs has been shown in a variety of in vivo models of ARDS, reducing bacterial pneumonia and ischemia-reperfusion injury while promoting the repair of ventilator-induced lung injury. This article reviews the current basic research findings and clinical applications of MSCs in the treatment of ARDS in order to emphasize the clinical prospects of MSCs.
Collapse
Affiliation(s)
- Tian-Yu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China
| | - Li-Hai Lu
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Si-Yu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zi-Hao Zheng
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai Shi
- Department of Respiratory Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Jing-Quan Liu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
13
|
Herrock O, Deer E, Amaral LM, Campbell N, Whitney D, Ingram N, Cornelius DC, Turner T, Hardy-Hardin J, Booz GW, Ibrahim T, LaMarca B. Inhibiting B cell activating factor attenuates preeclamptic symptoms in placental ischemic rats. Am J Reprod Immunol 2023; 89:e13693. [PMID: 36794639 PMCID: PMC10009902 DOI: 10.1111/aji.13693] [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: 11/28/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
PROBLEM Preeclampsia (PE), new-onset hypertension during pregnancy, is associated with a pro-inflammatory state with activated T cells, cytolytic natural killer (NK) cells, dysregulated complement proteins, and B cells secreting agonistic autoantibodies to the angiotensin II type-1 receptor (AT1-AA). The reduced uterine perfusion pressure (RUPP) model of placental ischemia recapitulates these features of PE. Blocking CD40L-CD40 communication between T and B cells or B cell depletion with Rituximab prevents hypertension and AT1-AA production in RUPP rats. This suggests that T cell-dependent B cell activation contributes to the hypertension and AT1-AA associated with PE. B2 cells maturing into antibody producing plasma cells are the product of T cell-dependent B cell-interactions and B cell Activating Factor (BAFF) is an integral cytokine in the development of B2 cells specifically. Thus, we hypothesize that BAFF blockade will selectively deplete B2 cells, therefore reducing blood pressure, AT1-AA, activated NK Cells, and complement in the RUPP rat model of PE. METHOD OF STUDY Gestational Day (GD) 14 pregnant rats underwent the RUPP procedure, and a subset were treated with 1 mg/kg Anti-BAFF antibodies via jugular catheters. On GD19, blood pressure was measured, B cells and NK cells were measured by flow cytometry, AT1-AA was measured by cardiomyocyte bioassay, and complement activation was measured by ELISA. RESULTS Anti-BAFF therapy attenuated hypertension, AT1-AA, NK cell activation, and APRIL levels in RUPP rats without negatively impacting fetal outcomes. CONCLUSIONS This study demonstrates that B2 cells contribute to hypertension, AT1-AA, and NK cell activation in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Lorena M. Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Darby Whitney
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Nicole Ingram
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | | | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Ja’Nasa Hardy-Hardin
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - George W. Booz
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Tarek Ibrahim
- 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
|
14
|
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
|
15
|
Mukherjee I, Singh S, Karmakar A, Kashyap N, Mridha AR, Sharma JB, Luthra K, Sharma RS, Biswas S, Dhar R, Karmakar S. New immune horizons in therapeutics and diagnostic approaches to Preeclampsia. Am J Reprod Immunol 2023; 89:e13670. [PMID: 36565013 DOI: 10.1111/aji.13670] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/02/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the commonest maladies, affecting 5%-10% of pregnancies worldwide. The American College of Obstetricians and Gynecologists (ACOG) identifies four categories of HDP, namely gestational hypertension (GH), Preeclampsia (PE), chronic hypertension (CH), and CH with superimposed PE. PE is a multisystem, heterogeneous disorder that encompasses 2%-8% of all pregnancy-related complications, contributing to about 9% to 26% of maternal deaths in low-income countries and 16% in high-income countries. These translate to 50 000 maternal deaths and over 500 000 fetal deaths worldwide, therefore demanding high priority in understanding clinical presentation, screening, diagnostic criteria, and effective management. PE is accompanied by uteroplacental insufficiency leading to vascular and metabolic changes, vasoconstriction, and end-organ ischemia. PE is diagnosed after 20 weeks of pregnancy in women who were previously normotensive or hypertensive. Besides shallow trophoblast invasion and inadequate remodeling of uterine arteries, dysregulation of the nonimmune system has been the focal point in PE. This results from aberrant immune system activation and imbalanced differentiation of T cells. Further, a failure of tolerance toward the semi-allogenic fetus results due to altered distribution of Tregs such as CD4+FoxP3+ or CD4+CD25+CD127(low) FoxP3+ cells, thereby creating a cytotoxic environment by suboptimal production of immunosuppressive cytokines like IL-10, IL-4, and IL-13. Also, intracellular production of complement protein C5a may result in decreased FoxP3+ regulatory T cells. With immune system dysfunction as a major driver in PE pathogenesis, it is logical that therapeutic targeting of components of the immune system with pharmacologic agents like anti-inflammatory and immune-modulating molecules are either being used or under clinical trial. Cholesterol synthesis inhibitors like Pravastatin may improve placental perfusion in PE, while Eculizumab (monoclonal antibody inhibiting C5) and small molecular inhibitor of C5a, Zilucoplan are under investigation. Monoclonal antibody against IL-17(Secukinumab) has been proposed to alter the Th imbalance in PE. Autologous Treg therapy and immune checkpoint inhibitors like anti-CTLA-4 are emerging as new candidates in immune horizons for PE management in the future.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.,Amity Institute of Biotechnology (AIB), Amity University, Noida, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Abhibrato Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asit Ranjan Mridha
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Radhey Shyam Sharma
- Ex-Head and Scientist G, Indian Council of Medical Research, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Noida, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Kang YE, Yi HS, Yeo MK, Kim JT, Park D, Jung Y, Kim OS, Lee SE, Kim JM, Joung KH, Lee JH, Ku BJ, Lee M, Kim HJ. Increased Pro-Inflammatory T Cells, Senescent T Cells, and Immune-Check Point Molecules in the Placentas of Patients With Gestational Diabetes Mellitus. J Korean Med Sci 2022; 37:e338. [PMID: 36513052 PMCID: PMC9745681 DOI: 10.3346/jkms.2022.37.e338] [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: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is the most common metabolic complication of pregnancy. To define the altered pathway in GDM placenta, we investigated the transcriptomic profiles from human placenta between GDM and controls. METHODS Clinical parameters and postpartum complications were reviewed in all participants. Differentially expressed canonical pathways were analyzed between the GDM and control groups based on transcriptomic analysis. CD4+ T, CD8+ T, and senescent T cell subsets were determined by flow cytometry based on staining for specific intracellular cytokines. RESULTS Gene ontology analysis revealed that the placenta of GDM revealed upregulation of diverse mitochondria or DNA replication related pathways and downregulation of T-cell immunity related pathways. The maternal placenta of the GDM group had a higher proportion of CD4+ T and CD8+ T cells than the control group. Interestingly, senescent CD4+ T cells tended to increase and CD8+ T cells were significantly increased in GDM compared to controls, along with increased programmed cell death-1 (CD274+) expression. Programmed death-ligand 1 expression in syncytotrophoblasts was also significantly increased in patients with GDM. CONCLUSION This study demonstrated increased proinflammatory T cells, senescent T cells and immune-check point molecules in GDM placentas, suggesting that changes in senescent T cells and immune-escape signaling might be related to the pathophysiology of GDM.
Collapse
Affiliation(s)
- Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyon-Seung Yi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung Tae Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Danbit Park
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Korea
| | - Yewon Jung
- Department of Obstetrics and Gynecology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ok Soon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seong Eun Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ji Min Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kyong Hye Joung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Bon Jeong Ku
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mina Lee
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Korea.
| | - Hyun Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea.
| |
Collapse
|
17
|
Akin S, Pinarbasi E, Bildirici AE, Cekin N. STOX1 promotor region -922 T > C polymorphism is associated with Early-Onset preeclampsia. J OBSTET GYNAECOL 2022; 42:3464-3470. [PMID: 36369889 DOI: 10.1080/01443615.2022.2141612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Preeclampsia (PE), affecting 5-8% of pregnancies, is a common pregnancy disease that has harmful effects on mother and foetus. It has been found that the STOX1 (Storkhead Box 1), which is a transcription factor, carries variants associated with PE. Previous studies showed that, there was a strong relationship between PE and STOX1 variants. Therefore, we hypothesised that variants in the promoter region of the gene may be related to the onset of PE. The aim of this study is to investigate the contribution of STOX1 gene promoter region variants to PE. The blood samples taken from 118 PE patients and 96 healthy pregnant women were analysed by Sanger sequencing method. Sequence analysis results showed that, there is a-922 T > C polymorphism (rs884181) in the promoter region of the STOX1 gene. This polymorphism was found to be statistically significant in individuals with early onset PE (p = 0.02) and in PE (p = 0.014) compared to the control group.IMPACT STATEMENTWhat is already known on this subject? As a result of whole-exon studies on the STOX1 gene, polymorphisms were found to disrupt the structure/expression/function of the gene and strengthen its relationship with PE and HELLP syndrome. A previous study by our team found an association between Y153H, the most common polymorphism of STOX1, and early onset PE.What do the results of this study add? In our study, it was aimed to investigate the effect of genetic modifications in STOX1 gene promoter region on PE through the maternal genotype. Because any change in the promoter region affects the expression level of the gene. Also, for the first time, sequence analysis of the promoter region of STOX1 is investigated in PE. The variations in STOX1 appear to be important in PE especially in Early Onset PE.What are the implications of these findings for clinical practice and/or further research? Although PE is a disease that occurs with pregnancy and shows its effects most during this period, women and children with a history of PE are more prone to various disorders, especially cardiovascular diseases in the following years. Therefore, understanding the pathogenesis of the disease is important for both prevention and treatment process. Variations on STOX1 appear to be important in terms of disease risk.
Collapse
Affiliation(s)
- Seyda Akin
- Faculty of Medicine, Department of Medical Biology, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Ergun Pinarbasi
- Faculty of Medicine, Department of Medical Biology, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Aslihan Esra Bildirici
- Faculty of Medicine, Department of Medical Biology, Sivas Cumhuriyet University, Sivas, Türkiye.,Exon Biyotek, Cumhuriyet Teknokent, Sivas, Türkiye
| | - Nilgun Cekin
- Faculty of Medicine, Department of Medical Biology, Sivas Cumhuriyet University, Sivas, Türkiye
| |
Collapse
|
18
|
Faulkner JL, Wright D, Antonova G, Jaffe IZ, Kennard S, Belin de Chantemèle EJ. Midgestation Leptin Infusion Induces Characteristics of Clinical Preeclampsia in Mice, Which Is Ablated by Endothelial Mineralocorticoid Receptor Deletion. Hypertension 2022; 79:1536-1547. [PMID: 35510543 DOI: 10.1161/hypertensionaha.121.18832] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Patients with preeclampsia demonstrate increases in placental leptin production in midgestation, and an associated increase in late gestation plasma leptin levels. The consequences of mid-late gestation increases in leptin production in pregnancy is unknown. Our previous work indicates that leptin infusion induces endothelial dysfunction in nonpregnant female mice via leptin-mediated aldosterone production and endothelial mineralocorticoid receptor (ECMR) activation, which is ablated by ECMR deletion. Therefore, we hypothesized that leptin infusion in mid-gestation of pregnancy induces endothelial dysfunction and hypertension, hallmarks of clinical preeclampsia, which are prevented by ECMR deletion. METHODS Leptin was infused via miniosmotic pump (0.9 mg/kg per day) into timed-pregnant ECMR-intact (WT) and littermate-mice with ECMR deletion (KO) on gestation day (GD)11-18. RESULTS Leptin infusion decreased fetal weight and placental efficiency in WT mice compared with WT+vehicle. Radiotelemetry recording demonstrated that blood pressure increased in leptin-infused WT mice during infusion. Leptin infusion reduced endothelial-dependent relaxation responses to acetylcholine (ACh) in both resistance (second-order mesenteric) and conduit (aorta) vessels in WT pregnant mice. Leptin infusion increased placental ET-1 (endothelin-1) production evidenced by increased PPET-1 (preproendothelin-1) and ECE-1 (endothelin-converting enzyme-1) expressions in WT mice. Adrenal aldosterone synthase (CYP11B2) and angiotensin II type 1 receptor b (AT1Rb) expression increased with leptin infusion in pregnant WT mice. KO pregnant mice demonstrated protection from leptin-induced reductions in pup weight, placental efficiency, increased BP, and endothelial dysfunction. CONCLUSIONS Collectively, these data indicate that leptin infusion in midgestation induces endothelial dysfunction, hypertension, and fetal growth restriction in pregnant mice, which is ablated by ECMR deletion.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Department of Physiology (J.L.F.), Medical College of Georgia at Augusta University
| | - Derrian Wright
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Galina Antonova
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.)
| | - Simone Kennard
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemèle
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University.,Department of Cardiology (E.J.B.d.C.), Medical College of Georgia at Augusta University
| |
Collapse
|
19
|
Regulatory T Cell Apoptosis during Preeclampsia May Be Prevented by Gal-2. Int J Mol Sci 2022; 23:ijms23031880. [PMID: 35163802 PMCID: PMC8836599 DOI: 10.3390/ijms23031880] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
There are several open questions to be answered regarding the pathophysiology of the development of preeclampsia (PE). Numerous factors are involved in its genesis, such as defective placentation, vascular impairment, and an altered immune response. The activation of the adaptive and innate immune system represents an immunologic, particularity during PE. Proinflammatory cytokines are predominantly produced, whereas immune regulatory and immune suppressive factors are diminished in PE. In the present study, we focused on the recruitment of regulatory T cells (Tregs) which are key players in processes mediating immune tolerance. To identify Tregs in the decidua, an immunohistochemical staining of FoxP3 of 32 PE and 34 control placentas was performed. A clearly reduced number of FoxP3-positive cells in the decidua of preeclamptic women could be shown in our analysis (p = 0.036). Furthermore, CCL22, a well-known Treg chemoattractant, was immunohistochemically evaluated. Interestingly, CCL22 expression was increased at the maternal-fetal interface in PE-affected pregnancies (psyncytiotrophoblast = 0.035, pdecidua = 0.004). Therefore, the hypothesis that Tregs undergo apoptosis at the materno-fetal interface during PE was generated, and verified by FoxP3/TUNEL (TdT-mediated dUTP-biotin nick end labeling) staining. Galectin-2 (Gal-2), a member of the family of carbohydrate-binding proteins, which is known to be downregulated during PE, seems to play a pivotal role in T cell apoptosis. By performing a cell culture experiment with isolated Tregs, we could identify Gal-2 as a factor that seems to prevent the apoptosis of Tregs. Our findings point to a cascade of apoptosis of Tregs at the materno-fetal interface during PE. Gal-2 might be a potential therapeutic target in PE to regulate immune tolerance.
Collapse
|
20
|
Bakrania BA, George EM, Granger JP. Animal models of preeclampsia: investigating pathophysiology and therapeutic targets. Am J Obstet Gynecol 2022; 226:S973-S987. [PMID: 33722383 PMCID: PMC8141071 DOI: 10.1016/j.ajog.2020.10.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/01/2020] [Accepted: 10/19/2020] [Indexed: 02/03/2023]
Abstract
Animal models have been critical in investigating the pathogenesis, mediators, and even therapeutic options for a number of diseases, including preeclampsia. Preeclampsia is the leading cause of maternal and fetal morbidity and mortality worldwide. The placenta is thought to play a central role in the pathogenesis of this disease because it releases antiangiogenic and proinflammatory factors into the maternal circulation, resulting in the maternal syndrome. Despite the deleterious effects preeclampsia has been shown to have on the mother and baby during pregnancy and postpartum, there is still no effective treatment for this disease. Although clinical studies in patients are crucial to identify the involvement of pathogenic factors in preeclampsia, there are obvious limitations that prevent detailed investigation of the quantitative importance of time-dependent mechanisms involved in this syndrome. Animal models allow investigators to perform proof-of-concept studies and examine whether certain factors found in women with preeclampsia mediate hypertension and other manifestations of this disease. In this brief review, we summarize some of the more widely studied models used to investigate pathophysiological mechanisms that are thought to be involved in preeclampsia. These include models of placental ischemia, angiogenic imbalance, and maternal immune activation. Infusion of preeclampsia-related factors into animals has been widely studied to understand the specific mediators of this disease. These models have been included, in addition to a number of genetic models involved in overexpression of the renin-angiotensin system, complement activation, and trophoblast differentiation. Together, these models cover multiple mechanisms of preeclampsia from trophoblast dysfunction and impaired placental vascularization to the excess circulating placental factors and clinical manifestation of this disease. Most animal studies have been performed in rats and mice; however, we have also incorporated nonhuman primate models in this review. Preclinical animal models not only have been instrumental in understanding the pathophysiology of preeclampsia but also continue to be important tools in the search for novel therapeutic options for the treatment of this disease.
Collapse
Affiliation(s)
- Bhavisha A Bakrania
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Eric M George
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Joey P Granger
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW It is well established that controlled immune activation and balance is critical for women's reproductive health and successful pregnancy outcomes. Research in recent decades in both clinical and animal studies has demonstrated that aberrant immune activation and inflammation play a role in the development and progression of women's reproductive health and pregnancy-related disorders. Inflammasomes are multi-protein cytoplasmic complexes that mediate immune activation. In this review, we summarize current knowledge on the role of inflammasome activation in pregnancy-related disorders. RECENT FINDINGS Increased activation of inflammasome is associated with multiple women's health reproductive disorders and pregnancy-associated disorders, including preeclampsia (PreE). Inflammasome activation is also associated with the novel coronavirus disease 2019 (COVID-19) disease caused by the SARS-Cov-2 virus. We and others have observed a positive association between increased PreE incidences with the onset of the COVID-19 pandemic. Here, we present our recent data indicating increased inflammasome activation, represented by caspase-1 activity, in women with COVID-19 and PreE compared to normotensive pregnant women COVID-19. The role of inflammation in pregnancy-related disorders is an area of intense research interest. With the onset of the COVID-19 pandemic and the associated increase in PreE observed clinically, there is a greater need to identify mechanisms of pathophysiology and targets to treat this maternal disorder. Inflammasome activation is associated with PreE and COVID-19 infection and may hold therapeutic potential to improve outcomes associated with PreE and curb the morbidity attributed to PreE.
Collapse
|
22
|
Ancuța E, Zamfir R, Martinescu G, Crauciuc DV, Ancuța C. The Complement System, T Cell Response, and Cytokine Shift in Normotensive versus Pre-Eclamptic and Lupus Pregnancy. J Clin Med 2021; 10:jcm10245722. [PMID: 34945017 PMCID: PMC8705505 DOI: 10.3390/jcm10245722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/14/2022] Open
Abstract
Successful pregnancy requires an immunological shift with T helper CD4+ bias based on disbalance Th1/Th17 versus Th2/T regulatory (Tregs) required to induce tolerance against the semi-allogeneic fetus and placenta and to support fetal growth. Considered a pregnancy-specific hypertensive disorder, pre-eclampsia is characterized by multifaceted organ involvement related to impaired maternal immune tolerance to paternal antigens triggered by hypoxic placental injury as well as excessive local and systemic anti-angiogenic and inflammatory factor synthesis. Both systemic and local Th1/Th2 shift further expands to Th17 cells and their cytokines (IL-17) complemented by suppressive Treg and Th2 cytokines (IL-10, IL-4); alterations in Th17 and Tregs cause hypertension during pregnancy throughout vasoactive factors and endothelial dysfunction, providing an explanatory link between immunological and vascular events in the pathobiology of pre-eclamptic pregnancy. Apart from immunological changes representative of normotensive pregnancy, lupus pregnancy is generally defined by higher serum pro-inflammatory cytokines, lower Th2 polarization, defective and lower number of Tregs, potential blockade of complement inhibitors by anti-phospholipid antibodies, and similar immune alterations to those seen in pre-eclampsia. The current review underpins the immune mechanisms of pre-eclampsia focusing on local (placental) and systemic (maternal) aberrant adaptive and innate immune response versus normotensive pregnancy and pregnancy in systemic autoimmune conditions, particularly lupus.
Collapse
Affiliation(s)
- Eugen Ancuța
- Research Department, “Elena Doamna” Obstetrics and Gynecology Clinical Hospital, 700398 Iași, Romania; (E.A.); (G.M.)
| | - Radu Zamfir
- Fundeni Clinical Institute, 022328 București, Romania;
| | - Gabriel Martinescu
- Research Department, “Elena Doamna” Obstetrics and Gynecology Clinical Hospital, 700398 Iași, Romania; (E.A.); (G.M.)
| | - Dragoș Valentin Crauciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Correspondence: (D.V.C.); or (C.A.); Tel.: +40-740036387 (C.A.)
| | - Codrina Ancuța
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- 2nd Rheumatology Department, Clinical Rehabilitation Hospital, 700661 Iași, Romania
- Correspondence: (D.V.C.); or (C.A.); Tel.: +40-740036387 (C.A.)
| |
Collapse
|
23
|
Dos Santos Fagundes I, Brendler EP, Nunes Erthal I, Eder Ribeiro RJ, Caron-Lienert RS, Machado DC, Pinheiro da Costa BE, Poli-de-Figueiredo CE. Total Th1/Th2 cytokines profile from peripheral blood lymphocytes in normal pregnancy and preeclampsia syndrome. Hypertens Pregnancy 2021; 41:15-22. [PMID: 34812111 DOI: 10.1080/10641955.2021.2008424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To evaluate total Th1/Th2 cytokines in CD3+ cells (immunocompetent T-lymphocytes) and peripheral blood lymphocytes, mostly CD4+ (T helper cells) and CD8+ (T-cytotoxic cells) subpopulations in preeclampsia. Total blood leukocytes and lymphocytes counts, percent cells: CD3+, INF-g+/CD3+, IL-4+/CD3+, and IL-10+/CD3+, CD4+/CD8+ were determined by flow-cytometry. Preeclampsia (n= 26) and normal pregnancy (n= 25) participants were age and gestational age matched. CD4+ lymphocytes count was higher in preeclampsia, compared with normal pregnancy (43.6 ± 5.8 vs 37.6 ± 5.6%; P< 0.001). CD3+ cells Th1/Th2 shift was not detected in preeclampsia, yet may be present in other cell types, such as CD4+ and CD3 - lymphocytes.
Collapse
Affiliation(s)
- Iara Dos Santos Fagundes
- Serviço de Imunologia Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Eduardo Pletsch Brendler
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | - Isadora Nunes Erthal
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | | | | | - Denise Cantarelli Machado
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | | | | |
Collapse
|
24
|
Booz GW, Kennedy D, Bowling M, Robinson T, Azubuike D, Fisher B, Brooks K, Chinthakuntla P, Hoang NH, Hosler JP, Cunningham MW. Angiotensin II type 1 receptor agonistic autoantibody blockade improves postpartum hypertension and cardiac mitochondrial function in rat model of preeclampsia. Biol Sex Differ 2021; 12:58. [PMID: 34727994 PMCID: PMC8562001 DOI: 10.1186/s13293-021-00396-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022] Open
Abstract
Women with preeclampsia (PE) have a greater risk of developing hypertension, cardiovascular disease (CVD), and renal disease later in life. Angiotensin II type I receptor agonistic autoantibodies (AT1-AAs) are elevated in women with PE during pregnancy and up to 2-year postpartum (PP), and in the reduced uterine perfusion pressure (RUPP) rat model of PE. Blockade of AT1-AA with a specific 7 amino acid peptide binding sequence (‘n7AAc’) improves pathophysiology observed in RUPP rats; however, the long-term effects of AT1-AA inhibition in PP is unknown. Pregnant Sprague Dawley rats were divided into three groups: normal pregnant (NP) (n = 16), RUPP (n = 15), and RUPP + ‘n7AAc’ (n = 16). Gestational day 14, RUPP surgery was performed and ‘n7AAc’ (144 μg/day) administered via osmotic minipump. At 10-week PP, mean arterial pressure (MAP), renal glomerular filtration rate (GFR) and cardiac functions, and cardiac mitochondria function were assessed. MAP was elevated PP in RUPP vs. NP (126 ± 4 vs. 116 ± 3 mmHg, p < 0.05), but was normalized in in RUPP + ‘n7AAc’ (109 ± 3 mmHg) vs. RUPP (p < 0.05). PP heart size was reduced by RUPP + ’n7AAc’ vs. RUPP rats (p < 0.05). Complex IV protein abundance and enzymatic activity, along with glutamate/malate-driven respiration (complexes I, III, and IV), were reduced in the heart of RUPP vs. NP rats which was prevented with ‘n7AAc’. AT1-AA inhibition during pregnancy not only improves blood pressure and pathophysiology of PE in rats during pregnancy, but also long-term changes in blood pressure, cardiac hypertrophy, and cardiac mitochondrial function PP.
Collapse
Affiliation(s)
- George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel Kennedy
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael Bowling
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Taprieka Robinson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel Azubuike
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Brandon Fisher
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Karen Brooks
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pooja Chinthakuntla
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ngoc H Hoang
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jonathan P Hosler
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mark W Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
25
|
Leelawai S, Suwanrath C, Pruphetkaew N, Chongphattararot P, Sathirapanya P. Gestational Bell's Palsy Is Associated with Higher Blood Pressure during Late Pregnancy and Lower Birth Weight: A Retrospective Case-Control Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph181910342. [PMID: 34639638 PMCID: PMC8507752 DOI: 10.3390/ijerph181910342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
The associations between gestational Bell's palsy (GBP) and late obstetric complications (LOCs), i.e., preeclampsia (PE), eclampsia (EC), gestational hypertension (GHT), and gestational diabetes mellitus (GDM) remain unclear. This study aimed to evaluate these associations and the neonatal health of the newborns born from pregnant women with and without GBP. A retrospective 1:5 case-control study matching exact maternal age and gravidity between pregnant women with and without GBP in Songklanagarind Hospital from 2006 to 2016 was conducted. The associations between GBP and PE, EC, GHT, and GDM, as well as comparison of the newborns' health indices were analyzed by bivariate analysis (p < 0.05). Eight GBP cases out of 8,756 pregnant women were recruited. Six GBP cases were first or second gravid. GBP occurred during the third trimester in five cases. Except for higher median systolic blood pressure (125 (114.2, 127.5) vs. (110 (107.0, 116.0), p = 0.045) and diastolic blood pressures (77 (73.0, 80.8) vs. 70 (65.0, 73.2), p = 0.021) in the GBP cases, associations between GBP and all LOCs could not be concluded due to the lack of power. However, a significantly lower mean birth weight in the newborns of GBP mothers was found (2672.2 (744.0) vs. 3154.8 (464.7), p = 0.016) with statistically significant power. Except for the higher blood pressures and lower birth weights of the newborns of GBP mothers, an association between GBP and LOCs remains inconclusive.
Collapse
Affiliation(s)
- Sumonthip Leelawai
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; (S.L.); (P.C.)
| | - Chitkasaem Suwanrath
- Department of Obstetrics and Gynecology, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand;
| | - Nannapat Pruphetkaew
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand;
| | - Pensri Chongphattararot
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; (S.L.); (P.C.)
| | - Pornchai Sathirapanya
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; (S.L.); (P.C.)
- Correspondence:
| |
Collapse
|
26
|
Gumusoglu S, Scroggins S, Vignato J, Santillan D, Santillan M. The Serotonin-Immune Axis in Preeclampsia. Curr Hypertens Rep 2021; 23:37. [PMID: 34351543 DOI: 10.1007/s11906-021-01155-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW To review the literature and detail the potential immune mechanisms by which hyperserotonemia may drive pro-inflammation in preeclampsia and to provide insights into potential avenues for therapeutic discovery. RECENT FINDINGS Preeclampsia is a severe hypertensive complication of pregnancy associated with significant maternal and fetal risk. Though it lacks any effective treatment aside from delivery of the fetus and placenta, recent work suggests that targeting serotonin systems may be one effective therapeutic avenue. Serotonin dysregulation underlies multiple domains of physiologic dysfunction in preeclampsia, including vascular hyporeactivity and excess platelet aggregation. Broadly, serotonin is increased across maternal and placental domains, driven by decreased catabolism and increased availability of tryptophan precursor. Pro-inflammation, another hallmark of the disease, may drive hyperserotonemia in preeclampsia. Interactions between immunologic dysfunction and hyperserotonemia in preeclampsia depend on multiple mechanisms, which we discuss in the present review. These include altered immune cell, kynurenine pathway metabolism, and aberrant cytokine production mechanisms, which we detail. Future work may leverage animal and in vitro models to reveal serotonin targets in the context of preeclampsia's immune biology, and ultimately to mitigate vascular and platelet dysfunction in the disease. Hyperserotonemia in preeclampsia drives pro-inflammation via metabolic, immune cell, and cytokine-based mechanisms. These immune mechanisms may be targeted to treat vascular and platelet endophenotypes in preeclampsia.
Collapse
Affiliation(s)
- Serena Gumusoglu
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
| | - Sabrina Scroggins
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Julie Vignato
- University of Iowa College of Nursing, Iowa City, Iowa, USA
| | - Donna Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Mark Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
27
|
Obesity-associated cardiovascular risk in women: hypertension and heart failure. Clin Sci (Lond) 2021; 135:1523-1544. [PMID: 34160010 DOI: 10.1042/cs20210384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
The pathogenesis of obesity-associated cardiovascular diseases begins long prior to the presentation of a cardiovascular event. In both men and women, cardiovascular events, and their associated hospitalizations and mortality, are often clinically predisposed by the presentation of a chronic cardiovascular risk factor. Obesity increases the risk of cardiovascular diseases in both sexes, however, the clinical prevalence of obesity, as well as its contribution to crucial cardiovascular risk factors is dependent on sex. The mechanisms via which obesity leads to cardiovascular risk is also discrepant in women between their premenopausal, pregnancy and postmenopausal phases of life. Emerging data indicate that at all reproductive statuses and ages, the presentation of a cardiovascular event in obese women is strongly associated with hypertension and its subsequent chronic risk factor, heart failure with preserved ejection fraction (HFpEF). In addition, emerging evidence indicates that obesity increases the risk of both hypertension and heart failure in pregnancy. This review will summarize clinical and experimental data on the female-specific prevalence and mechanisms of hypertension and heart failure in women across reproductive stages and highlight the particular risks in pregnancy as well as emerging data in a high-risk ethnicity in women of African ancestry (AA).
Collapse
|
28
|
Ishimwe JA. Maternal microbiome in preeclampsia pathophysiology and implications on offspring health. Physiol Rep 2021; 9:e14875. [PMID: 34042284 PMCID: PMC8157769 DOI: 10.14814/phy2.14875] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
Preeclampsia is a devastating hypertensive pregnancy disorder that currently affects 2%–8% of pregnancies worldwide. It is associated with maternal and fetal mortality and morbidity and adverse health outcomes both in mom and offspring beyond pregnancy. The pathophysiology is not completely understood, and there are no approved therapies to specifically treat for the disease, with only few therapies approved to manage symptoms. Recent advances suggest that aberrations in the composition of the microbiome may play a role in the pathogenesis of various diseases including preeclampsia. The maternal and uteroplacental environments greatly influence the long‐term health outcomes of the offspring through developmental programming mechanisms. The current review summarizes recent developments on the role of the microbiome in adverse pregnancy outcomes with a focus on preeclampsia. It also discusses the potential role of the maternal microbiome in fetal programming; explores gut‐targeted therapeutics advancement and their implications in the treatment of preeclampsia.
Collapse
Affiliation(s)
- Jeanne A Ishimwe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
29
|
Collier ARY, Smith LA, Karumanchi SA. Review of the immune mechanisms of preeclampsia and the potential of immune modulating therapy. Hum Immunol 2021; 82:362-370. [PMID: 33551128 DOI: 10.1016/j.humimm.2021.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
Successful pregnancy relies on maternal immunologic tolerance mechanisms limit maladaptive immune responses against the semi-allogeneic fetus and placenta and support fetal growth. Preeclampsia is a common disorder of pregnancy that affects 4-10% of pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Preeclampsia clinically manifests as maternal hypertension, proteinuria, and progressive multi-organ injury likely triggered by hypoxic injury to the placenta, resulting in local and systemic anti-angiogenic and inflammatory factor production. Despite the steady rising rates of preeclampsia in the United States, effective treatment options are limited to delivery, which improves maternal status often at the cost of prematurity in the newborn. Preeclampsia also increases the lifelong risk of cardiovascular disease for both mother and infant. Thus, identifying new therapeutic targets is a high priority area to improve maternal, fetal, and infant health outcomes. Immune abnormalities in the placenta and in the maternal circulation have been reported to precede the clinical onset of disease. In particular, excessive systemic and placental complement activation and impaired adaptive T cell tolerance with Th1/Th2/Th17/Treg imbalance has been reported in humans and in animal models of preeclampsia. In this review, we focus on the evidence for the immune origins of preeclampsia, discuss the promise of immune modulating therapy for prevention or treatment, and highlight key areas for future research.
Collapse
Affiliation(s)
- Ai-Ris Y Collier
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA; Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Laura A Smith
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - S Ananth Karumanchi
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
30
|
Cunningham MW, Amaral LM, Campbell NE, Cornelius DC, Ibrahim T, Vaka VR, LaMarca B. Investigation of interleukin-2-mediated changes in blood pressure, fetal growth restriction, and innate immune activation in normal pregnant rats and in a preclinical rat model of preeclampsia. Biol Sex Differ 2021; 12:4. [PMID: 33407826 PMCID: PMC7789596 DOI: 10.1186/s13293-020-00345-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/06/2020] [Indexed: 01/16/2023] Open
Abstract
Two important clinical features of preeclampsia (PE) are hypertension and fetal growth restriction. The reduced uterine perfusion pressure (RUPP) preclinical rat model of PE exhibits both of these features. Moreover, RUPP and PE women have elevated vasoconstrictor peptide endothelin-1 (ET-1) and inflammation. Interleukin-2 (IL-2) is a cytokine that regulates NK cell activity and is elevated in miscarriage, PE, and RUPP rats. The objective of this study was to examine a role for IL-2 in NK cell activation, fetal growth restriction, and hypertension during pregnancy by either infusion of IL-2 or blockade of IL-2 (basiliximab) in normal pregnant (NP) and RUPP rats. On gestational day 14, NP and RUPP rats received low (LD), middle (MD), or high dose (HD) IL-2 (0.05, 0.10, or 0.20 ng/ml) IP or basiliximab (0.07 mg per rat) by IV infusion. On day 19, blood pressure (MAP), pup weights, and blood were collected. Basiliximab had no effect on blood pressure, however, significantly lowered NK cells and may have worsened overall fetal survival in RUPP rats. However, IL-2 LD (102 ± 4 mmHg) and IL-2 HD (105 ± 6 mmHg) significantly lowered blood pressure, ET-1, and activated NK cells compared to control RUPPs (124 ± 3 mmHg, p < 0.05). Importantly, IL-2 in RUPP rats significantly reduced fetal weight and survival. These data indicate that although maternal benefits may have occurred with low dose IL-2 infusion, negative effects were seen in the fetus. Moreover, inhibition of IL-2 signaling did not have favorable outcome for the mother or fetus.
Collapse
Affiliation(s)
- Mark W. Cunningham
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Lorena M. Amaral
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Nathan E. Campbell
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Denise C. Cornelius
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
- Department Of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS USA
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Venkata Ramana Vaka
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, Center for Excellence in Renal and Cardiovascular Research, University of Mississippi Medical Center, Jackson, MS 39216 USA
- Division of Maternal Fetal Medicine, Department Of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS 39216 USA
| |
Collapse
|
31
|
Deer E, Reeve KE, Amaral L, Vaka VR, Franks M, Campbell N, Fitzgerald S, Herrock O, Ibrahim T, Cornelius D, LaMarca B. CD4+ T cells cause renal and placental mitochondrial oxidative stress as mechanisms of hypertension in response to placental ischemia. Am J Physiol Renal Physiol 2021; 320:F47-F54. [PMID: 33196321 PMCID: PMC7847053 DOI: 10.1152/ajprenal.00398.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
The reduced uterine perfusion pressure (RUPP) rat model and normal pregnant (NP) rat recipients of RUPP CD4+ T cells recapitulate many characteristics of preeclampsia such as hypertension and oxidative stress. We have shown an important hypertensive role for natural killer (NK) cells to cause mitochondrial dysfunction in RUPP rats; however, the role for RUPP CD4+ T cells to stimulate NK cells is unknown. Therefore, we hypothesized that RUPP-induced CD4+ T cells activate NK cells to cause mitochondrial dysfunction/reactive oxygen species (ROS) as mechanisms of hypertension during pregnancy. We tested our hypothesis by adoptive transfer of RUPP CD4+ T cells into NP rats or by inhibiting the activation of RUPP CD4+ T cells with Orencia (abatacept) and examining hypertension, NK cells, and mitochondrial function. RUPP was performed on gestation day (GD) 14, and splenic CD4+ T cells were isolated on GD 19 and injected into NP rats on GD 13. In a separate group of rats, Orencia was infused and the RUPP procedure was performed. Mean arterial pressure and placental and renal mitochondrial ROS increased in RUPP (n = 7, P < 0.05) and NP + RUPP CD4+ T-cell recipients (n = 13, P < 0.05) compared with control NP (n = 7) and NP + NP CD4+ T-cell recipients (n = 5) but was reduced with Orencia (n = 13, P < 0.05). Placental and renal respiration was reduced in RUPP (n = 6, P < 0.05) and NP + RUPP CD4+ T-cell recipients (n = 6, state 3 P < 0.05) compared with NP (n = 5) and NP + NP CD4+ T-cell recipients (n = 5) but improved with Orencia (n = 9, n = 8 P < 0.05). These data indicate that CD4+ T cells, independent of NK cells, cause mitochondrial dysfunction/ROS contributing to hypertension in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kristin E Reeve
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department Of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Venkata Ramana Vaka
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael Franks
- 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
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Owen Herrock
- 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
| | - Denise Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department Of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - 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
|
32
|
Wang Y, Li Z, Song G, Wang J. Potential of Immune-Related Genes as Biomarkers for Diagnosis and Subtype Classification of Preeclampsia. Front Genet 2020; 11:579709. [PMID: 33335538 PMCID: PMC7737719 DOI: 10.3389/fgene.2020.579709] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/30/2020] [Indexed: 11/13/2022] Open
Abstract
Objective Preeclampsia is the main cause of maternal mortality due to a lack of diagnostic biomarkers and effective prevention and treatment. The immune system plays an important role in the occurrence and development of preeclampsia. This research aimed to identify significant immune-related genes to predict preeclampsia and possible prevention and control methods. Methods Differential expression analysis between normotensive and PE pregnancies was performed to identify significantly changed immune-related genes. Generalized linear model (GLM), random forest (RF), and support vector machine (SVM) models were established separately to screen the most suitable biomarkers for the diagnosis of PE among these significantly changed immune-related genes. The consensus clustering method was used to divide the PE cases into several subgroups to explore the function of the significantly changed immune-related genes in PE. Results Thirteen significantly changed immune-related genes were obtained by the differential expression analysis. RF was the best model and was used to select the four most important explanatory variables (CRH, PI3, CCL18, and CCL2) to diagnose PE. A nomogram model was constructed to predict PE based on these four variables. The decision curve analysis (DCA) and clinical impact curves revealed that PE patients could significantly benefit from this nomogram. Consensus clustering analysis of the 13 differentially expressed immune-related genes (DIRGs) was used to identify 3 subgroups of PE pregnancies with different clinical outcomes and immune cell infiltration. Conclusion Our study identified four immune-related genes to predict PE and three subgroups of PE with different clinical outcomes and immune cell infiltration. Future studies on the three subgroups may provide direction for individualized treatment of PE patients.
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhen Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guiyu Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
33
|
Leelawai S, Sathirapanya P, Suwanrath C. Bell's Palsy in Pregnancy: A Case Series. Case Rep Neurol 2020; 12:452-459. [PMID: 33442374 PMCID: PMC7772825 DOI: 10.1159/000509682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/21/2020] [Indexed: 11/19/2022] Open
Abstract
The association between pregnancy-associated Bell's palsy (PABP) and gestational hypertension (GHT), preeclampsia (PE), and eclampsia (EC) remains inconclusive. We aimed to study the characteristics of PABP cases and the neonatal outcomes at our institution. All cases diagnosed with PABP from 2006 to 2016 were identified. Demographic and clinical characteristics including maternal age, previous medical and obstetric illnesses, gestational age at the onset of PABP, the development of PE/EC, GHT, gestational diabetes mellitus (GDM), treatment and outcomes, as well as neonatal health indices and anomalies were described. Eight patients with PABP were identified. Most of the cases were first- or second-gravidity pregnancies. PABP occurred during the third trimester except for one case in whom PABP developed 2 days postpartum. No PABP case associated with EC was found. PE was found in only one case in whom GHT occurred in a previous pregnancy. Moreover, GHT combined with GDM was found in a case with previous GHT. The recovery of PABP was satisfactory. Previous obstetric complications are associated with the current PE, GHT and GDM. Facial weakness recovers favorably regardless of treatment and the neonatal outcomes are overall satisfactory.
Collapse
Affiliation(s)
- Sumonthip Leelawai
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Pornchai Sathirapanya
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Chitkasaem Suwanrath
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
34
|
Muyayalo KP, Huang DH, Zhao SJ, Xie T, Mor G, Liao AH. COVID-19 and Treg/Th17 imbalance: Potential relationship to pregnancy outcomes. Am J Reprod Immunol 2020; 84:e13304. [PMID: 32662111 PMCID: PMC7404618 DOI: 10.1111/aji.13304] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/27/2022] Open
Abstract
Caused by a novel type of virus, severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), coronavirus disease 2019 (COVID‐19) constitutes a global public health emergency. Pregnant women are considered to have a higher risk of severe morbidity and even mortality due to their susceptibility to respiratory pathogens and their particular immunologic state. Several studies assessing SARS‐CoV‐2 infection during pregnancy reported adverse pregnancy outcomes in patients with severe conditions, including spontaneous abortion, preterm labor, fetal distress, cesarean section, preterm birth, neonatal asphyxia, neonatal pneumonia, stillbirth, and neonatal death. However, whether these complications are causally related to SARS‐CoV‐2 infection is not clear. Here, we reviewed the scientific evidence supporting the contributing role of Treg/Th17 cell imbalance in the uncontrolled systemic inflammation characterizing severe cases of COVID‐19. Based on the recognized harmful effects of these CD4+ T‐cell subset imbalances in pregnancy, we speculated that SARS‐CoV‐2 infection might lead to adverse pregnancy outcomes through the deregulation of otherwise tightly regulated Treg/Th17 ratios, and to subsequent uncontrolled systemic inflammation. Moreover, we discuss the possibility of vertical transmission of COVID‐19 from infected mothers to their infants, which could also explain adverse perinatal outcomes. Rigorous monitoring of pregnancies and appropriate measures should be taken to prevent and treat early eventual maternal and perinatal complications.
Collapse
Affiliation(s)
- Kahinho P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Dong-Hui Huang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Jia Zhao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Xie
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Cunningham MW, Jayaram A, Deer E, Amaral LM, Vaka VR, Ibrahim T, Cornelius DC, LaMarca B. Tumor necrosis factor alpha (TNF-α) blockade improves natural killer cell (NK) activation, hypertension, and mitochondrial oxidative stress in a preclinical rat model of preeclampsia. Hypertens Pregnancy 2020; 39:399-404. [PMID: 32646252 DOI: 10.1080/10641955.2020.1793999] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The RUPP rat model of Preeclampsia exhibits hypertension (MAP), cytolytic natural killer (cNK) cells, tumor necrosis factor alpha (TNF-α) and mitochondrial Reactive Oxygen Species (mt ROS). Objective: Does TNF-α blockade with ETAN (Etanercept) decrease cNK cell and mt ROS in RUPP rats. METHODS On gestational day 14, RUPP surgery was performed, ETAN (0.4 mg/kg) was administered on day 18, MAP, blood and tissues collected on 19. RESULTS MAP, cytolytic NK cells and mt ROS were elevated in RUPP vs. NP and normalized with ETAN. CONCLUSION TNF-α blockade lowered blood pressure and improve inflammation and organ function in response to placental ischemia.
Collapse
Affiliation(s)
- Mark W Cunningham
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Aswathi Jayaram
- 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
| | - Evangeline Deer
- 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
| | - Venkata Ramana Vaka
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Denise C Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center , Jackson, MS, USA.,Department of Emergency Medicine, 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
|
36
|
Turbeville HR, Sasser JM. Preeclampsia beyond pregnancy: long-term consequences for mother and child. Am J Physiol Renal Physiol 2020; 318:F1315-F1326. [PMID: 32249616 PMCID: PMC7311709 DOI: 10.1152/ajprenal.00071.2020] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 01/26/2023] Open
Abstract
Preeclampsia is defined as new-onset hypertension after the 20th wk of gestation along with evidence of maternal organ failure. Rates of preeclampsia have steadily increased over the past 30 yr, affecting ∼4% of pregnancies in the United States and causing a high economic burden (22, 69). The pathogenesis is multifactorial, with acknowledged contributions by placental, vascular, renal, and immunological dysfunction. Treatment is limited, commonly using symptomatic management and/or early delivery of the fetus (6). Along with significant peripartum morbidity and mortality, current research continues to demonstrate that the consequences of preeclampsia extend far beyond preterm delivery. It has lasting effects for both mother and child, resulting in increased susceptibility to hypertension and chronic kidney disease (45, 54, 115, 116), yielding lifelong risk to both individuals. This review discusses recent guideline updates and recommendations along with current research on these long-term consequences of preeclampsia.
Collapse
Affiliation(s)
- Hannah R Turbeville
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer M Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
37
|
Li M, Qian L, Yu J, Zou Y. Interleukin-35 inhibits human umbilical vein endothelial cell injury induced by sera from pre-eclampsia patients by up-regulating S100A8 protein expression. Hypertens Pregnancy 2020; 39:126-138. [PMID: 32200685 DOI: 10.1080/10641955.2020.1744000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objectives: The protective effects of interleukin(IL)-35 against injury to human umbilical vein endothelial cells (HUVECs) induced by the serum of pre-eclampsia patients were analyzed.Methods: This cross-sectional study included 24 patients with pre-eclampsia (PE) and 24 normotensive pregnant women.Results: Compared to normotensive pregnant women, patients with pre-eclampsia had lower IL-35 levels (P < 0.05). In addition, our in vitro experiments, IL-35 inhibited the PE serum-induced apoptosis of HUVECs and the levels of reactive oxygen species in HUVECs.Conclusion: Decreases in the serum IL-35 level may play an important role in the pathogenesis of endothelial dysfunction in patients with pre-eclampsia.
Collapse
Affiliation(s)
- Ming Li
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, Jiangsu Province, P. R. China
| | - Lei Qian
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, Jiangsu Province, P. R. China
| | - Jianxiu Yu
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, Jiangsu Province, P. R. China
| | - Yingfen Zou
- Department of Obstetrics and Gynecology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Jiangsu Province, P. R. China
| |
Collapse
|
38
|
Zhu A, Reeder SB, Johnson KM, Nguyen SM, Fain SB, Bird IM, Golos TG, Wieben O, Shah DM, Hernando D. Quantitative ferumoxytol-enhanced MRI in pregnancy: A feasibility study in the nonhuman primate. Magn Reson Imaging 2020; 65:100-108. [PMID: 31655139 PMCID: PMC6956847 DOI: 10.1016/j.mri.2019.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To assess the feasibility of ferumoxytol-enhanced MRI in pregnancy with a nonhuman primate model. MATERIALS AND METHODS In this prospective study, eleven pregnant rhesus macaques at day 98 ± 5 of gestation were divided into three groups, untreated control (UC) (n = 3), saline control (SC) (n = 4) and interleukin 1 beta (IL-1β) treated (IT) (n = 4), which were administered with either saline or IL-1β into the amniotic fluid. All animals were imaged at multiple time points before and after ferumoxytol administration (4 mg/kg). Longitudinal R2* and susceptibility of tissues were obtained using region-of-interest analysis and the longitudinal changes were assessed using linear mixed models and Student's t-test. RESULTS In fetuses, a slope of 0.3 s-1/day (P = 0.008), 0.00 ppm/day (P = 0.699) and - 0.2 s-1/day (P = 0.023) was observed in liver R2*, liver susceptibility, and lung R2*, respectively. In placentas, R2* and susceptibility increased immediately after ferumoxytol administration (P < 0.001) and decreased to baseline within two days. The mean change from baseline showed no significant difference between the SC group and the IT group at all scan time points. In maternal livers, R2* increased immediately after ferumoxytol administration, further increased at one-day, and then decreased but remained elevated (P < 0.001). The mean change from baseline showed no significant difference between the SC group and the IT group at all scan time points. CONCLUSIONS This work demonstrates the feasibility of quantitative ferumoxytol-enhanced MRI to measure dynamics of ferumoxytol delivery and washout in the placenta. Stable MRI measurements indicated no evidence of iron deposition in fetal tissues of nonhuman primates after maternal ferumoxytol exposure.
Collapse
Affiliation(s)
- Ante Zhu
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Scott B Reeder
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Department of Radiology, University of Wisconsin, Madison, WI, USA; Department of Medical Physics, University of Wisconsin, Madison, WI, USA; Department of Medicine, University of Wisconsin, Madison, WI, USA; Department of Emergency Medicine, University of Wisconsin, Madison, WI, USA
| | - Kevin M Johnson
- Department of Radiology, University of Wisconsin, Madison, WI, USA; Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Sydney M Nguyen
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA; Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
| | - Sean B Fain
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Department of Radiology, University of Wisconsin, Madison, WI, USA; Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Ian M Bird
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA; Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Oliver Wieben
- Department of Radiology, University of Wisconsin, Madison, WI, USA; Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Dinesh M Shah
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
| | - Diego Hernando
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Department of Radiology, University of Wisconsin, Madison, WI, USA; Department of Medical Physics, University of Wisconsin, Madison, WI, USA; Department of Electrical and Computer Engineering, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
39
|
Sarı İ, Ökten H, Aktan Ç, Cihan E. Association of the sEH gene promoter polymorphisms and haplotypes with preeclampsia. J Med Biochem 2020; 39:428-435. [PMID: 33312058 DOI: 10.5937/jomb0-27745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Background The epoxyeicosatrienoic acids (EETs) have antihypertensive, anti-inflammatory, and organ protective properties and their circulation levels are related to hypertension, diabetes mellitus, cardiovascular diseases, and preeclampsia. Soluble epoxide hydrolase (sEH) catalyses the degradation of EETs to less biologically active dihydroxyeicosatrienoic acids. Here, we sequenced the promoter region of EPHX2 to investigate the association between promoter sequence alterations that we thought to affect the expression levels of the enzyme and preeclampsia (PE). Methods Nucleotide sequencing of the promoter region of the EPHX2, spanning from position -671 to +30, was performed on 100 pregnant women with PE and, 20 or more weeks pregnant normotensive, healthy women (n=100). Results Pregnant women who carry rs4149235, rs4149232, rs73227309, and rs62504268 polymorphisms have 4.4, 2.4, 2.3, and 2.8 times significantly increased risk of PE, respectively. CCGG (OR: 3.11; 95% CI: 1.12-8.62) and CCCA (OR: 0.45; 95% CI: 0.36-0.55) haplotypes were associated with an increased and decreased risk of PE, respectively. Conclusions Four SNPs (rs4149232, rs4149235, rs73227309, and rs62504268) in the promoter region of the EPHX2, and CCGG and CCCA haplotypes of these 4 SNPs were significantly associated with PE. These SNPs in the promoter region may affect sEH expression and thus enzyme activity and may play a role in PE pathogenesis by causing individual differences in EET levels. However, future studies are needed to confirm our findings and examine the effect of these SNPs on the sEH expression and/or enzyme activity.
Collapse
Affiliation(s)
- İsmail Sarı
- Niğde Omer Halisdemir University, Faculty of Medicine, Department of Medical Biochemistry, Niğde, Turkey
| | - Hatice Ökten
- Beykent University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Çağdaş Aktan
- Beykent University, Faculty of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Esra Cihan
- Niğde Omer Halisdemir University, Faculty of Medicine, Department of Obstetrics and Gynaecology, Niğde, Turkey
| |
Collapse
|
40
|
Vaka VR, Cunningham MW, Deer E, Franks M, Ibrahim T, Amaral LM, Usry N, Cornelius DC, Dechend R, Wallukat G, LaMarca BD. Blockade of endogenous angiotensin II type I receptor agonistic autoantibody activity improves mitochondrial reactive oxygen species and hypertension in a rat model of preeclampsia. Am J Physiol Regul Integr Comp Physiol 2019; 318:R256-R262. [PMID: 31721604 DOI: 10.1152/ajpregu.00179.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Preeclampsia (PE) is characterized by new-onset hypertension that usually occurs in the third trimester of pregnancy and is associated with oxidative stress and angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs). Inhibition of the AT1-AAs in the reduced uterine perfusion pressure (RUPP) rat, a model of PE, attenuates hypertension and many other characteristics of PE. We have previously shown that mitochondrial oxidative stress (mtROS) is a newly described PE characteristic exhibited by the RUPP rat that contributes to hypertension. However, the factors that cause mtROS in PE or RUPP are unknown. Thus, the objective of the current study is to use pharmacologic inhibition of AT1-AAs to examine their role in mtROS in the RUPP rat model of PE. AT1-AA inhibition in RUPP rats was achieved by administration of an epitope-binding peptide ('n7AAc'). Female Sprague-Dawley rats were divided into the following two groups: RUPP and RUPP + AT1-AA inhibition (RUPP + 'n7AAc'). On day 14 of gestation (GD), RUPP surgery was performed; 'n7AAc' peptide (2 µg/μL) was administered by miniosmotic pumps in a subset of RUPP rats; and on GD19, sera, placentas, and kidneys were collected. mitochondrial respiration and mtROS were measured in isolated mitochondria using the Oxygraph 2K and fluorescent microplate reader, respectively. Placental and renal mitochondrial respiration and mtROS were improved in RUPP + 'n7AAc' rats compared with RUPP controls. Moreover, endothelial cells (human umbilical vein endothelial cells) treated with RUPP + 'n7AAc' sera exhibited less mtROS compared with those treated with RUPP sera. Overall, our findings suggest that AT1-AA signaling is one stimulus of mtROS during PE.
Collapse
Affiliation(s)
- Venkata Ramana Vaka
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mark W Cunningham
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael Franks
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nathan Usry
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Berlin, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center, Charité Campus Buch, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Babbette D LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Obstetrics & Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
41
|
Rabaglino MB, Conrad KP. Evidence for shared molecular pathways of dysregulated decidualization in preeclampsia and endometrial disorders revealed by microarray data integration. FASEB J 2019; 33:11682-11695. [PMID: 31356122 DOI: 10.1096/fj.201900662r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microarray data of chorionic villous samples (CVSs) obtained from women of ∼11.5 gestational weeks who developed preeclampsia with severe features (sPE; PE-CVS) revealed a molecular signature of impaired endometrial maturation (decidualization) before and during early pregnancy. Because endometrial disorders are also associated with aberrant decidualization, we asked whether they share molecular features with sPE. We employed microarray data integration to compare the molecular pathologies of PE-CVS and endometrial disorders, as well as decidua obtained postpartum from women with sPE. Eight public databases were reanalyzed with R software to determine differentially expressed genes (DEGs) in pathologic tissues relative to normal controls. DEGs were then compared to explore overlap. Shared DEGs were examined for enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Principal component and network analyses were subsequently applied to selected DEGs. There was significant overlap of DEGs changing in the same direction for PE-CVS and endometrial disorders, suggesting common molecular pathways. Shared DEGs were enriched for cytokine-cytokine receptor interaction. Genes in this pathway revealed expression patterns forming 2 distinct clusters, one for normal and the other pathologic endometrium. The most affected hub genes were related to decidualization and NK cell function. Few DEGs were shared by PE-CVS, and PE decidua obtained postpartum. sPE may be part of a biologic continuum of "endometrial spectrum disorders."-Rabaglino, M. B., Conrad, K. P. Evidence for shared molecular pathways of dysregulated decidualization in preeclampsia and endometrial disorders revealed by microarray data integration.
Collapse
Affiliation(s)
- Maria Belen Rabaglino
- Instituto de Investigación en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina; and
| | - Kirk P Conrad
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA.,Department of Obstetrics and Gynecology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
42
|
Anto EO, Roberts P, Coall D, Turpin CA, Adua E, Wang Y, Wang W. Integration of suboptimal health status evaluation as a criterion for prediction of preeclampsia is strongly recommended for healthcare management in pregnancy: a prospective cohort study in a Ghanaian population. EPMA J 2019; 10:211-226. [PMID: 31462939 DOI: 10.1007/s13167-019-00183-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Abstract
Background Normotensive pregnancy may develop into preeclampsia (PE) and other adverse pregnancy complications (APCs), for which the causes are still unknown. Suboptimal health status (SHS), a physical state between health and disease, might contribute to the development and progression of PE. By integration of a routine health measure in this Ghanaian Suboptimal Health Cohort Study, we explored the usefulness of a 25-question item SHS questionnaire (SHSQ-25) for early screening and prediction of normotensive pregnant women (NTN-PW) likely to develop PE. Methods We assessed the overall health status among a cohort of 593 NTN-PW at baseline (10-20 weeks gestation) and followed them at 21-31 weeks until 32-42 weeks. After an average of 20 weeks follow-up, 498 participants returned and were included in the final analysis. Hematobiochemical, clinical and sociodemographic data were obtained. Results Of the 498 participants, 49.8% (248/498) had 'high SHS' at baseline (61.7% (153/248) later developed PE) and 38.3% (95/248) were NTN-PW, whereas 50.2% (250/498) had 'optimal health' (17.6% (44/250) later developed PE) and 82.4% (206/250) were NTN-PW. At baseline, high SHS score yielded a significantly (p < 0.05) increased adjusted odds ratio, a wider area under the curve (AUC) and a higher sensitivity and specificity for the prediction of PE (3.67; 0.898; 91.9% and 87.8%), PE coexisting with intrauterine growth restriction (2.86, 0.838; 91.5% and 75.9%), stillbirth (2.52; 0.783; 96.6% and 60.0%), hemolysis elevated liver enzymes and low platelet count (HELLP) syndrome (2.08; 0.800; 97.2% and 63.8%), acute kidney injury (2.20; 0.825; 95.3% and 70.0%) and dyslipidaemia (2.80; 0.8205; 95.7% and 68.4%) at 32-42 weeks gestation. Conclusions High SHS score is associated with increased incidence of PE; hence, SHSQ-25 can be used independently as a risk stratification tool for adverse pregnancy outcomes thereby creating an opportunity for predictive, preventive and personalized medicine.
Collapse
Affiliation(s)
- Enoch Odame Anto
- 1School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia.,2Department of Molecular Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Peter Roberts
- 1School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | - David Coall
- 1School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | | | - Eric Adua
- 1School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia
| | - Youxin Wang
- 4Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Wei Wang
- 1School of Medical and Health Sciences, Edith Cowan University, Perth, WA Australia.,4Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China.,5School of Public Health, Taishan Medical University, Taian, China
| |
Collapse
|
43
|
Muyayalo KP, Huang X, Qian Z, Li Z, Mor G, Liao A. Low circulating levels of vitamin D may contribute to the occurrence of preeclampsia through deregulation of Treg /Th17 cell ratio. Am J Reprod Immunol 2019; 82:e13168. [DOI: 10.1111/aji.13168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Kahindo P. Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiao‐Bo Huang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhu Qian
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zhi‐Hui Li
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- C.S. Mott Center for Human Growth and Development Wayne State University School of Medicine Detroit MI USA
| | - Ai‐Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
44
|
Downregulation of α7 nicotinic acetylcholine receptors in peripheral blood monocytes is associated with enhanced inflammation in preeclampsia. BMC Pregnancy Childbirth 2019; 19:188. [PMID: 31138166 PMCID: PMC6540389 DOI: 10.1186/s12884-019-2340-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/13/2019] [Indexed: 11/18/2022] Open
Abstract
Background Preeclampsia is associated with chronic inflammation. The cholinergic anti-inflammatory pathway regulates systemic inflammation through activating α7 nicotinic acetylcholine receptors (α7nAChR) expressed in monocytes/macrophages. This study aimed to investigate the role of α7nAChR in peripheral blood monocytes in preeclampsia. Methods Peripheral blood monocytes were isolated from 30 nonpregnant (NP), 32 normotensive pregnant (NT), and 35 preeclamptic (PE) women. Results We found that both protein and mRNA expression levels of α7nAChR in monocytes from the PE women were significantly lower than those of the NP and NT women (both p < 0.01). α7nAChR protein expression levels in monocytes were negatively correlated with levels of systolic blood pressure (r = − 0.40, p = 0.04), proteinuria (r = − 0.54, p < 0.01), tumor necrosis factor-alpha (TNF-α, r = − 0.42, p = 0.01), and interleukin (IL)-1β (r = − 0.56, p < 0.01), while positively correlated with IL-10 levels (r = 0.43, p = 0.01) in the PE women. Both baseline and lipopolysaccharides (LPS)-induced increase of TNF-α, IL-1β, and IL-6 levels from monocytes were higher in the PE group than the NP and NT groups (all p < 0.01), but IL-10 levels in the PE group was lower than that of the NP and NT groups (p < 0.01). In addition, the NF-κB activity in monocytes from the PE women was higher than the NP and NT women (p < 0.01). Importantly, activation of α7nAChR with its agonist PNU-282987 inhibited NF-κB, decreased TNF-α, IL-1β, and IL-6 release, and increased IL-10 release in monocytes from the PE women (all p < 0.01). Conclusion In conclusion, these findings suggest that downregulation of α7nAChR may be associated with the development of preeclampsia through increasing pro-inflammatory and decreasing anti-inflammatory cytokine release via the NF-κB pathway.
Collapse
|
45
|
Travis OK, White D, Pierce WA, Ge Y, Stubbs CY, Spradley FT, Williams JM, Cornelius DC. Chronic infusion of interleukin-17 promotes hypertension, activation of cytolytic natural killer cells, and vascular dysfunction in pregnant rats. Physiol Rep 2019; 7:e14038. [PMID: 30963715 PMCID: PMC6453821 DOI: 10.14814/phy2.14038] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 01/06/2023] Open
Abstract
Previous studies by our lab have established that placental-ischemia stimulated T-helper 17 cells (TH 17s) cause increased cytolytic natural killer (cNK) cell proliferation and activation during pregnancy; however, the exact mechanism is unknown. The objective of this study was to investigate the role of interlukin 17 (IL-17) in inducing cNK cell activation in pregnancy. We infused 150 pg/day of recombinant IL-17 into a subset of normal pregnant (NP) Sprague Dawley rats from gestation day (GD) 12-19 (NP+IL-17). On GD 19, mean arterial pressure (MAP), fetal and placental weights, cytokines, cNK cell activation, cytotoxic enzymes, and vascular reactivity were assessed. MAP significantly increased from 99 ± 3 mmHg in NP to 120 ± 1 mmHg in NP+IL-17 (P < 0.05). Fetal weight significantly decreased from 2.52 ± 0.04 g in NP to 2.32 ± 0.03 g in NP+IL-17 as did placental weight (NP: 0.65 ± 0.03 g; NP+IL-17: 0.54 ± 0.01 g, P < 0.05). Plasma levels of TNF-α increased to 281.4 ± 55.07 pg/mL in NP+IL-17 from 145.3 ± 16.03 pg/mL in NP (P < 0.05) while placental levels of VEGF decreased from 74.2 ± 6.48 pg/mg in NP to 54.2 ± 3.19 pg/mg in NP+IL-17. Total NK cells were increased in the placenta (NP: 14.3 ± 3.49%; NP+IL-17: 29.33 ± 2.76%, P < 0.05) as were cytolytic NK cells (NP: 3.31 ± 1.25%; NP+IL-17: 13.41 ± 1.81%, P < 0.05). A similar trend was observed in circulating NK cells. Plasma granzyme K increased from 3.55 ± 2.29 pg/mL in NP to 20.9 ± 7.76 pg/mL in NP+IL-17 (P < 0.05), and plasma granzyme B increased from 10.95 ± 0.64 pg/mL in NP to 14.9 ± 0.98 pg/mL in NP+IL-17(P < 0.05). In the placenta, both granzyme A (NP: 246.1 ± 16.7 pg/mg; NP+IL-17: 324.3 ± 15.07 pg/mg, P < 0.05) and granzyme B (NP: 15.18 ± 3.79 pg/mg; NP+IL-17: 27.25 ± 2.34 pg/mg, P < 0.05) increased in response to IL-17 infusion. Finally, vascular reactivity of uterine arteries was significantly impaired in response to IL-17 infusion. The results of this study suggest that IL-17 plays a significant role in the activation of cNK cells during pregnancy.
Collapse
Affiliation(s)
- Olivia K. Travis
- Department of Experimental Therapeutics and PharmacologyogyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Dakota White
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| | - W. Austin Pierce
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| | - Ying Ge
- Department of SurgeryUniversity of Mississippi Medical CenterJacksonMississippi
| | - Cassandra Y. Stubbs
- Department of Experimental Therapeutics and PharmacologyogyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Frank T. Spradley
- Department of SurgeryUniversity of Mississippi Medical CenterJacksonMississippi
| | - Jan M. Williams
- Department of Experimental Therapeutics and PharmacologyogyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Denise C. Cornelius
- Department of Experimental Therapeutics and PharmacologyogyUniversity of Mississippi Medical CenterJacksonMississippi
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| |
Collapse
|
46
|
Kelley AS, Banker M, Goodrich JM, Dolinoy DC, Burant C, Domino SE, Smith YR, Song PXK, Padmanabhan V. Early pregnancy exposure to endocrine disrupting chemical mixtures are associated with inflammatory changes in maternal and neonatal circulation. Sci Rep 2019; 9:5422. [PMID: 30931951 PMCID: PMC6443771 DOI: 10.1038/s41598-019-41134-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/26/2019] [Indexed: 01/12/2023] Open
Abstract
Endocrine disrupting chemicals (EDCs) are ubiquitous, and pregnancy is a sensitive window for toxicant exposure. EDCs may disrupt the maternal immune system, which may lead to poor pregnancy outcomes. Most studies investigate single EDCs, even though "real life" exposures do not occur in isolation. We tested the hypothesis that uniquely weighted mixtures of early pregnancy exposures are associated with distinct changes in the maternal and neonatal inflammasome. First trimester urine samples were tested for 12 phthalates, 12 phenols, and 17 metals in 56 women. Twelve cytokines were measured in first trimester and term maternal plasma, and in cord blood after delivery. Spearman correlations and linear regression were used to relate individual exposures with inflammatory cytokines. Linear regression was used to relate cytokine levels with gestational age and birth weight. Principal component analysis was used to assess the effect of weighted EDC mixtures on maternal and neonatal inflammation. Our results demonstrated that maternal and cord blood cytokines were differentially associated with (1) individual EDCs and (2) EDC mixtures. Several individual cytokines were positively associated with gestational age and birth weight. These observed associations between EDC mixtures and the pregnancy inflammasome may have clinical and public health implications for women of childbearing age.
Collapse
Affiliation(s)
- Angela S Kelley
- Department of Obstetrics and Gynecology, University of Michigan, L4001 Women's Hospital, 1500 East Medical Center Drive, Ann Arbor, Michigan, 48109, USA
| | - Margaret Banker
- Department of Biostatistics, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA.,Department of Nutritional Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA
| | - Charles Burant
- Department of Internal Medicine, University of Michigan, 24 Frank Lloyd Wright Drive, Ann Arbor, Michigan, 48105, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan, L4001 Women's Hospital, 1500 East Medical Center Drive, Ann Arbor, Michigan, 48109, USA
| | - Yolanda R Smith
- Department of Obstetrics and Gynecology, University of Michigan, L4001 Women's Hospital, 1500 East Medical Center Drive, Ann Arbor, Michigan, 48109, USA
| | - Peter X K Song
- Department of Biostatistics, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, L4001 Women's Hospital, 1500 East Medical Center Drive, Ann Arbor, Michigan, 48109, USA. .,Department of Environmental Health Sciences, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, Michigan, 48109, USA. .,Department of Pediatrics, University of Michigan, 7510 MSRB 1, 1500 W. Medical Center Dr., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
47
|
Sljivancanin Jakovljevic T, Kontic-Vucinic O, Nikolic N, Carkic J, Soldatovic I, Milasin J. Glutathione-S-transferase M1 polymorphism and pro-inflammatory cytokines tumour necrosis factor-α and interleukin-1β are associated with preeclampsia in Serbian women. Am J Reprod Immunol 2019; 81:e13105. [PMID: 30811718 DOI: 10.1111/aji.13105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/23/2019] [Accepted: 02/18/2019] [Indexed: 12/30/2022] Open
Abstract
PROBLEM Preeclampsia has a multifactorial origin with genetic, immunological, and environmental factors described as main contributors to its onset. This study aimed to investigate glutathione-S-transferase M1 (GSTM1) and glutathione-S-transferase T1 (GSTT1) gene polymorphisms, the expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), and the potential relationship between GST polymorphisms and cytokine expression levels in preeclampsia and uncomplicated pregnancy. METHOD OF STUDY This prospective case-control study included 50 women with preeclampsia and 50 healthy pregnant women. DNA and RNA were extracted from women leukocytes. Deletion polymorphisms were analyzed by PCR, while cytokine mRNA expression was analyzed by real-time PCR. RESULTS GSTM1 null genotype with present GSTT1 increased the risk for preeclampsia development. Deletion of GSTT1 without deletion of GSTM1 increased the risk for early preeclampsia. Relative mRNA expression of TNF-α was significantly higher in preeclampsia compared to healthy pregnant women (P = 0.006). Expression of IL-1β was significantly higher in severe and late preeclampsia compared to the control group (P = 0.005, P = 0.007, respectively). A significant positive correlation between TNF-α and IL-1β was observed (Spearman's ρ = 0.312, P = 0.028) and between IL-1β and IL-6, in preeclampsia group (Spearman's ρ = 0.296, P = 0.037). IL-1β was significantly increased in patients with GSTT1 null genotype (P = 0.015) while IL-6 was increased in patients with GSTM1 null genotype (P = 0.015). CONCLUSIONS GSTM1 null genotype represents a risk factor for preeclampsia development, while GSTT1 null genotype favors early preeclampsia. Preeclampsia is also associated with increased expression of pro-inflammatory cytokines, predominantly TNF-α and IL-1β.
Collapse
Affiliation(s)
| | - Olivera Kontic-Vucinic
- Department of Human Reproduction, Clinic of Obstetrics and Gynecology, Clinical Center of Serbia, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nadja Nikolic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Carkic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivan Soldatovic
- Institute of Medical Statistics and Informatics, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Milasin
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
48
|
Robertson SA, Green ES, Care AS, Moldenhauer LM, Prins JR, Hull ML, Barry SC, Dekker G. Therapeutic Potential of Regulatory T Cells in Preeclampsia-Opportunities and Challenges. Front Immunol 2019; 10:478. [PMID: 30984163 PMCID: PMC6448013 DOI: 10.3389/fimmu.2019.00478] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation is a central feature and is implicated as a causal factor in preeclampsia and other hypertensive disorders of pregnancy. Inflammatory mediators and leukocytes, which are elevated in peripheral blood and gestational tissues, contribute to the uterine vascular anomalies and compromised placental function that characterize particularly the severe, early onset form of disease. Regulatory T (Treg) cells are central mediators of pregnancy tolerance and direct other immune cells to counteract inflammation and promote robust placentation. Treg cells are commonly perturbed in preeclampsia, and there is evidence Treg cell insufficiency predates onset of symptoms. A causal role is implied by mouse studies showing sufficient numbers of functionally competent Treg cells must be present in the uterus from conception, to support maternal vascular adaptation and prevent later placental inflammatory pathology. Treg cells may therefore provide a tractable target for both preventative strategies and treatment interventions in preeclampsia. Steps to boost Treg cell activity require investigation and could be incorporated into pregnancy planning and preconception care. Pharmacological interventions developed to target Treg cells in autoimmune conditions warrant consideration for evaluation, utilizing rigorous clinical trial methodology, and ensuring safety is paramount. Emerging cell therapy tools involving in vitro Treg cell generation and/or expansion may in time become relevant. The success of preventative and therapeutic approaches will depend on resolving several challenges including developing informative diagnostic tests for Treg cell activity applicable before conception or during early pregnancy, selection of relevant patient subgroups, and identification of appropriate windows of gestation for intervention.
Collapse
Affiliation(s)
- Sarah A. Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Ella S. Green
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alison S. Care
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Lachlan M. Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - M. Louise Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Women's and Children's Hospital, Adelaide, SA, Australia
| | - Simon C. Barry
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Gustaaf Dekker
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
49
|
Turbeville HR, Taylor EB, Garrett MR, Didion SP, Ryan MJ, Sasser JM. Superimposed Preeclampsia Exacerbates Postpartum Renal Injury Despite Lack of Long-Term Blood Pressure Difference in the Dahl Salt-Sensitive Rat. Hypertension 2019; 73:650-658. [PMID: 30612494 PMCID: PMC6374193 DOI: 10.1161/hypertensionaha.118.12097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Preeclampsia results in increased susceptibility to hypertension and chronic kidney disease postpartum; however, the mechanisms responsible for disease progression in these women remain unknown. The purpose of this study was to test the hypothesis that 2 mechanisms contribute to the link between the maternal syndrome of preeclampsia and the increased postpartum risk of cardiovascular and renal disease: (1) increased T cells in the kidney and (2) a decreased NO:ET-1 (endothelin-1) ratio. Dahl S rats (a previously characterized model of preeclampsia superimposed on chronic hypertension) who experienced 2 pregnancies and virgin littermate controls were studied at 6 months of age. Mean arterial pressure was measured via telemetry, and renal injury was assessed through both histological analysis and measurement of urinary markers including nephrin, podocalyxin, and KIM-1 (kidney injury marker 1). Contributing mechanisms were assessed through flow cytometric analysis of renal T cells, quantification of plasma TNF-α (tumor necrosis factor-α) and IL-10 (interleukin-10), and quantification of urinary concentrations of NO metabolites and ET-1. Although prior pregnancy did not exacerbate the hypertension at 6 months, this group showed greater renal injury compared with virgin littermates. Flow cytometric analyses revealed an increase in renal T cells after pregnancy, and cytokine analysis revealed a systemic proinflammatory shift. Finally, the NO:ET-1 ratio was reduced. These results demonstrate that the link between the maternal syndrome of superimposed preeclampsia and postpartum risk of chronic kidney disease could involve both immune system activation and dysregulation of the NO:ET-1 balance.
Collapse
Affiliation(s)
- Hannah R. Turbeville
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Erin B. Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R. Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sean P. Didion
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael J. Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer M. Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
50
|
Cunningham MW, Vaka VR, McMaster K, Ibrahim T, Cornelius DC, Amaral L, Campbell N, Wallukat G, McDuffy S, Usry N, Dechend R, LaMarca B. Renal natural killer cell activation and mitochondrial oxidative stress; new mechanisms in AT1-AA mediated hypertensive pregnancy. Pregnancy Hypertens 2018; 15:72-77. [PMID: 30825931 DOI: 10.1016/j.preghy.2018.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022]
Abstract
Women with preeclampsia (PE) have increased mean arterial pressure (MAP), natural killer (NK) cells, reactive oxygen species (ROS), and agonistic autoantibodies to the angiotensin II type 1 receptor (AT1-AA). AT1-AA's administered to pregnant rodents produces a well-accepted model of PE. However, the role of NK cells and mitochondrial reactive oxygen species (mtROS) in AT1-AA mediated hypertension during pregnancy is unknown. We hypothesize that AT1-AA induced model of PE will exhibit elevated MAP, NK cells, and mtROS; while inhibition of the AT1-AA binding to the AT1R would be preventative. Pregnant rats were divided into 4 groups: normal pregnant (NP) (n = 5), NP + AT1-AA inhibitory peptide (NP +'n7AAc') (n = 3), NP + AT1-AA infused (NP + AT1-AA) (n = 10), and NP + AT1-AA +'n7AAc' (n = 8). Day 13, rats were surgically implanted with mini-pumps infusing either AT1-AA or AT1-AA +'n7AAc'. Day 19, tissue and blood was collected. MAP was elevated in AT1-AA vs. NP (119 ± 1 vs. 102 ± 2 mmHg, p < 0.05) and this was prevented by 'n7AAc' (108 ± 3). There was a 6 fold increase in renal activated NK cells in AT1-AA vs NP (1.2 ± 0.4 vs. 0.2 ± 0.1% Gated, p = 0.05) which returned to NP levels in AT1-AA +'n7AAc' (0.1 ± 0.1% Gated). Renal mtROS (317 ± 49 vs. 101 ± 13% Fold, p < 0.05) was elevated with AT1-AA vs NP and was decreased in AT1-AA +'n7AAc' (128 ± 16, p < 0.05). In conclusion, AT1-AA's increased MAP, NK cells, and mtROS which were attenuated by AT1-AA inhibition, thus highlighting new mechanisms of AT1-AA and the importance of drug therapy targeted to AT1-AAs in hypertensive pregnancies.
Collapse
Affiliation(s)
- Mark W Cunningham
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Venkata Ramana Vaka
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kristen McMaster
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Tarek Ibrahim
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Denise C Cornelius
- Depart. of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lorena Amaral
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Nathan Campbell
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Gerd Wallukat
- Experimental and Clinical Research Center, Charité Campus Buch, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Shyanne McDuffy
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Nathan Usry
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Berlin, Germany
| | - Babbette LaMarca
- Depart. of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States; Depart. of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States.
| |
Collapse
|