1
|
Warrington JP, Collins HE, Davidge ST, do Carmo JM, Goulopoulou S, Intapad S, Loria AS, Sones JL, Wold LE, Zinkhan EK, Alexander BT. Guidelines for in vivo models of developmental programming of cardiovascular disease risk. Am J Physiol Heart Circ Physiol 2024; 327:H221-H241. [PMID: 38819382 PMCID: PMC11380980 DOI: 10.1152/ajpheart.00060.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Research using animals depends on the generation of offspring for use in experiments or for the maintenance of animal colonies. Although not considered by all, several different factors preceding and during pregnancy, as well as during lactation, can program various characteristics in the offspring. Here, we present the most common models of developmental programming of cardiovascular outcomes, important considerations for study design, and provide guidelines for producing and reporting rigorous and reproducible cardiovascular studies in offspring exposed to normal conditions or developmental insult. These guidelines provide considerations for the selection of the appropriate animal model and factors that should be reported to increase rigor and reproducibility while ensuring transparent reporting of methods and results.
Collapse
Grants
- 20YVNR35490079 American Heart Association (AHA)
- R01HL139348 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135158 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54GM115428 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01AG057046 HHS | NIH | National Institute on Aging (NIA)
- P20 GM104357 NIGMS NIH HHS
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 GM149404 NIGMS NIH HHS
- P20GM104357 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM135002 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL163003 NHLBI NIH HHS
- R01HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01DK121411 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Excellence Faculty Support Grant Jewish Heritage Fund
- P30GM149404 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P30GM14940 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- 23SFRNPCS1067044 American Heart Association (AHA)
- R01 HL146562 NHLBI NIH HHS
- R56HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54 GM115428 NIGMS NIH HHS
- 1R01HL163076 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL51971 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- FS154313 CIHR
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Helen E Collins
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, Kentucky, United States
| | - Sandra T Davidge
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jussara M do Carmo
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, United States
- Department of Gynecology, and Obstetrics, Loma Linda University, Loma Linda, California, United States
| | - Suttira Intapad
- Department of Pharmacology, Tulane University, New Orleans, Louisiana, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jenny L Sones
- Equine Reproduction Laboratory, Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Erin K Zinkhan
- Department of Pediatrics, University of Utah and Intermountain Health, Salt Lake City, Utah, United States
- Intermountain Health, Salt Lake City, Utah, United States
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
2
|
van Kammen CM, Taal SEL, Wever KE, Granger JP, Lely AT, Terstappen F. Reduced uterine perfusion pressure as a model for preeclampsia and fetal growth restriction in murine: a systematic review and meta-analysis. Am J Physiol Heart Circ Physiol 2024; 327:H89-H107. [PMID: 38758122 PMCID: PMC11380978 DOI: 10.1152/ajpheart.00056.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The reduced uterine perfusion pressure (RUPP) model is frequently used to study preeclampsia and fetal growth restriction. An improved understanding of influential factors might improve reproducibility and reduce animal use considering the variability in RUPP phenotype. We performed a systematic review and meta-analysis by searching Medline and Embase (until 28 March, 2023) for RUPP studies in murine. Primary outcomes included maternal blood pressure (BP) or proteinuria, fetal weight or crown-rump length, fetal reabsorptions, or antiangiogenic factors. We aimed to identify influential factors by meta-regression analysis. We included 155 studies. Our meta-analysis showed that the RUPP procedure results in significantly higher BP (MD = 24.1 mmHg; [22.6; 25.7]; n = 148), proteinuria (SMD = 2.3; [0.9; 3.8]; n = 28), fetal reabsorptions (MD = 50.4%; [45.5; 55.2]; n = 42), circulating soluble FMS-like tyrosine kinase-1 (sFlt-1) (SMD = 2.6; [1.7; 3.4]; n = 34), and lower fetal weight (MD = -0.4 g; [-0.47; -0.34]; n = 113. The heterogeneity (variability between studies) in primary outcomes appeared ≥90%. Our meta-regression identified influential factors in the method and time point of BP measurement, randomization in fetal weight, and type of control group in sFlt-1. The RUPP is a robust model considering the evident differences in maternal and fetal outcomes. The high heterogeneity reflects the observed variability in phenotype. Because of underreporting, we observed reporting bias and a high risk of bias. We recommend standardizing study design by optimal time point and method chosen for readout measures to limit the variability. This contributes to improved reproducibility and thereby eventually improves the translational value of the RUPP model.
Collapse
Affiliation(s)
- Caren M van Kammen
- Division of Nanomedicine, Department CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Seija E L Taal
- Department of Woman and Baby, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Kimberley E Wever
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joey P Granger
- Department of Physiology and Biophysics, Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - A Titia Lely
- Department of Woman and Baby, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Fieke Terstappen
- Department of Woman and Baby, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
3
|
Wang X, Shields C, Tardo G, Peacock G, Hester E, Anderson M, Williams JM, Cornelius DC. IL-33 supplementation improves uterine artery resistance and maternal hypertension in response to placental ischemia. Am J Physiol Heart Circ Physiol 2024; 326:H1006-H1016. [PMID: 38363211 PMCID: PMC11279736 DOI: 10.1152/ajpheart.00045.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
Preeclampsia (PE), a leading cause of maternal/fetal morbidity and mortality, is a hypertensive pregnancy disorder with end-organ damage that manifests after 20 wk of gestation. PE is characterized by chronic immune activation and endothelial dysfunction. Clinical studies report reduced IL-33 signaling in PE. We use the Reduced Uterine Perfusion Pressure (RUPP) rat model, which mimics many PE characteristics including reduced IL-33, to identify mechanisms mediating PE pathophysiology. We hypothesized that IL-33 supplementation would improve blood pressure (BP), inflammation, and oxidative stress (ROS) during placental ischemia. We implanted intraperitoneal mini-osmotic pumps infusing recombinant rat IL-33 (1 µg/kg/day) into normal pregnant (NP) and RUPP rats from gestation day 14 to 19. We found that IL-33 supplementation in RUPP rats reduces maternal blood pressure and improves the uterine artery resistance index (UARI). In addition to physiological improvements, we found decreased circulating and placental cytolytic Natural Killer cells (cNKs) and decreased circulating, placental, and renal TH17s in IL-33-treated RUPP rats. cNK cell cytotoxic activity also decreased in IL-33-supplemented RUPP rats. Furthermore, renal ROS and placental preproendothelin-1 (PPET-1) decreased in RUPP rats treated with IL-33. These findings demonstrate a role for IL-33 in controlling vascular function and maternal BP during pregnancy by decreasing inflammation, renal ROS, and PPET-1 expression. These data suggest that IL-33 may have therapeutic potential in managing PE.NEW & NOTEWORTHY Though decreased IL-33 signaling has been clinically associated with PE, the mechanisms linking this signaling pathway to overall disease pathophysiology are not well understood. This study provides compelling evidence that mechanistically links reduced IL-33 with the inflammatory response and vascular dysfunction observed in response to placental ischemia, such as in PE. Data presented in this study submit the IL-33 signaling pathway as a possible therapeutic target for the treatment of PE.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Corbin Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Geilda Tardo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Greg Peacock
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emily Hester
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Marissa Anderson
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
4
|
Rodrigues SD, da Silva MLS, Martins LZ, Gomes SEB, Mariani NAP, Silva EJR, Kushima H, Mattos BR, Rizzi E, Dias-Junior CA. Pregnancy hypertension-associated endothelial dysfunction is attenuated by isoflurane anesthesia: Evidence of protective effect related to increases in nitric oxide. Life Sci 2023; 331:122039. [PMID: 37648198 DOI: 10.1016/j.lfs.2023.122039] [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: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
AIMS Pregnancy hypertension-induced endothelial dysfunction associated with impairment of nitric oxide (NO) bioavailability and hemodynamic derangements is a challenging for urgent procedures requiring maternal anesthesia. The volatile anesthetic isoflurane has demonstrated NO-associated protective effects. However, this isoflurane-induced effect is still unclear in pregnancy hypertension. Therefore, the present study examined the potential protective effects of isoflurane anesthesia on endothelial dysfunction and hemodynamic changes induced by hypertensive pregnancy associated with fetal and placental growth restrictions. MATERIALS AND METHODS Animals were distributed into four groups: normotensive pregnant rats (Preg), anesthetized pregnant rats (Preg+Iso), hypertensive pregnant rats (HTN-Preg), and anesthetized hypertensive pregnant rats (HTN-Preg+Iso). Systolic and diastolic pressures, mean arterial pressure (MAP), heart rate, fetal and placental weights, vascular contraction, endothelium-derived NO-dependent vasodilation, and NO levels were assessed. The vascular endothelial growth factor (VEGF) levels and endothelial NO synthase (eNOS) Serine (1177) phosphorylation (p-eNOS) expression were also examined. KEY FINDINGS Isoflurane produced more expressive hypotensive effects in the HTN-Preg+Iso versus Preg+Iso group, with respective reductions in MAP by 50 ± 13 versus 25 ± 4 mmHg (P < 0.05). Also, HTN-Preg+Iso compared to the HTN-Preg group showed (respectively) preventions against the weight loss of the fetuses (4.0 ± 0.6 versus 2.8 ± 0.6 g, P < 0.05) and placentas (0.37 ± 0.06 versus 0.30 ± 0.06 mg, P < 0.05), hyper-reactive vasocontraction response (1.8 ± 0.4 versus 2.8 ± 0.6 g, P < 0.05), impaired endothelium-derived NO-dependent vasodilation (84 ± 8 versus 50 ± 17 %, P < 0.05), reduced VEGF levels (147 ± 46 versus 25 ± 13 pg/mL, P < 0.05), and decreased p-eNOS expression (0.24 ± 0.07 versus 0.09 ± 0.05 arbitrary units, P < 0.05). SIGNIFICANCE Isoflurane anesthesia protects maternal endothelial function in pregnancy hypertension, and possibly endothelium-derived NO is involved.
Collapse
Affiliation(s)
- Serginara David Rodrigues
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Maria Luiza Santos da Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Laisla Zanetoni Martins
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Sáskia Estela Biasotti Gomes
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Noemia A P Mariani
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Erick J R Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Hélio Kushima
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil
| | - Bruna Rahal Mattos
- Unit of Biotechnology, University of Ribeirao Preto (UNAERP), Ribeirao Preto 14096-900, SP, Brazil
| | - Elen Rizzi
- Unit of Biotechnology, University of Ribeirao Preto (UNAERP), Ribeirao Preto 14096-900, SP, Brazil
| | - Carlos Alan Dias-Junior
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, 18618-689, SP, Brazil.
| |
Collapse
|
5
|
Alkhatib B, Salimi S, Jabari M, Padmanabhan V, Vyas AK. Impact of Adverse Gestational Milieu on Maternal Cardiovascular Health. Endocrinology 2023; 164:bqad060. [PMID: 37042476 PMCID: PMC10164662 DOI: 10.1210/endocr/bqad060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/13/2023]
Abstract
Cardiovascular disease affects 1% to 4% of the nearly 4 million pregnancies in the United States each year and is the primary cause of pregnancy-related mortality. Adverse pregnancy outcomes are associated with cardiovascular complications during pregnancy persisting into the postpartum period. Recently, investigations have identified an altered sex hormone milieu, such as in the case of hyperandrogenism, as a causative factor in the development of gestational cardiovascular dysfunction. The mechanisms involved in the development of cardiovascular disease in postpartum women are largely unknown. Animal studies have attempted to recapitulate adverse pregnancy outcomes to investigate causal relationships and molecular underpinnings of adverse gestational cardiac events and progression to the development of cardiovascular disease postpartum. This review will focus on summarizing clinical and animal studies detailing the impact of adverse pregnancy outcomes, including preeclampsia, gestational diabetes mellitus, and maternal obesity, on gestational cardiometabolic dysfunction and postpartum cardiovascular disease. Specifically, we will highlight the adverse impact of gestational hyperandrogenism and its potential to serve as a biomarker for maternal gestational and postpartum cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St. Louis, MO 63110, USA
| | - Shadi Salimi
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Mary Jabari
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | | | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St. Louis, MO 63110, USA
- College of Human Medicine, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
6
|
Wang X, Travis OK, Shields CA, Tardo GA, Giachelli C, Nutter CW, Glenn HL, Cooper OG, Davis T, Thomas R, Williams JM, Cornelius DC. NLRP3 inhibition improves maternal hypertension, inflammation, and vascular dysfunction in response to placental ischemia. Am J Physiol Regul Integr Comp Physiol 2023; 324:R556-R567. [PMID: 36847598 PMCID: PMC10069976 DOI: 10.1152/ajpregu.00192.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/01/2023]
Abstract
Preeclampsia (PE) is a pregnancy-specific hypertensive disorder with end-organ damage that presents after 20 wk of gestation. PE pathophysiology often includes vascular dysfunction and increased inflammation that continues to damage patient health even after PE resolves. Currently, there is no cure for PE beyond delivery of the fetal-placental unit. Previous clinical studies have identified elevated placental NLRP3 expression in patients with PE and suggest NLRP3 as a potential therapeutic target. In this study, we examined the effect of NLRP3 inhibition on PE pathophysiology in the reduced uterine perfusion pressure (RUPP) model rat using MCC950 (20 mg/kg/day) or esomeprazole (3.5 mg/kg/day). We hypothesized that increased NLRP3 in response to placental ischemia impairs anti-inflammatory IL-33 signaling to induce T-helper 17 cell (TH17) and cytolytic NK cell (cNK) activation, which is known to mediate oxidative stress and vascular dysfunction leading to maternal HTN and intrauterine growth restriction. RUPP rats had significantly higher placental NLRP3 expression, maternal blood pressure, fetal reabsorption rate, vascular resistance, oxidative stress, cNKs and TH17s, and decreased IL-33 compared with normal pregnant (NP) rats. NLRP3 inhibition, with either treatment, significantly reduced placental NLRP3 expression, maternal blood pressure, fetal reabsorption rates, vascular resistance, oxidative stress, cNK, and TH17 populations in RUPP rats. Based on our findings, NLRP3 inhibition reduces PE pathophysiology and esomeprazole may be a potential therapeutic for PE treatment.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Olivia K Travis
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Corbin A Shields
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - G Ann Tardo
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Chelsea Giachelli
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Christopher W Nutter
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Hannah L Glenn
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Olive G Cooper
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Tatiana Davis
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Rashauna Thomas
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
7
|
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
|
8
|
Herrock OT, Deer E, Amaral LM, Campbell N, Lemon J, Ingram N, Cornelius DC, Turner TW, Fitzgerald S, Ibrahim T, Dechend R, Wallukat G, LaMarca B. B2 cells contribute to hypertension and natural killer cell activation possibly via AT1-AA in response to placental ischemia. Am J Physiol Renal Physiol 2023; 324:F179-F192. [PMID: 36417275 PMCID: PMC9844978 DOI: 10.1152/ajprenal.00190.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia, new onset hypertension during pregnancy, is associated with activated T helper cells (Th) and B cells secreting agonistic autoantibodies against the angiotensin II type 1 receptor (AT1-AA). The reduced uterine perfusion pressure (RUPP) model of placental ischemia recapitulates these characteristics. We have shown that Th-B cell communication contributes to AT1-AA and symptoms of preeclampsia in the RUPP rat. B2 cells are classical B cells that communicate with Th cells and are then transformed into memory B cells. We hypothesize that B2 cells cause hypertension, natural killer (NK) cell activation, and complement activation during pregnancy through the production of AT1-AA. To test this hypothesis, total splenic B cells and B2 cells were isolated from normal pregnant (NP) or RUPP rats on gestational day (GD)19 and adoptively transferred into GD12 NP rats. A group of recipient rats was treated with a specific inhibitor peptide of AT1-AA. On GD19, mean arterial pressure was measured, tissues were collected, activated NK cells were measured by flow cytometry, and AT1-AA was measured by cardiomyocyte assay. NP recipients of RUPP B cells or RUPP B2 cells had increased mean arterial pressure, AT1-AA, and circulating activated NK cells compared with recipients of NP B cells. Hypertension in NP recipients of RUPP B cells or RUPP B2 was attenuated with AT1-AA blockade. This study demonstrates that B cells and B2 cells from RUPP rats cause hypertension and increased AT1-AA and NK cell activation in response to placental ischemia during pregnancy.NEW & NOTEWORTHY This study demonstrates that placental ischemia-stimulated B2 cells induce hypertension and circulating natural killer cell activation and angiotensin II type 1 receptor production in normal pregnant rats.
Collapse
Affiliation(s)
- Owen T Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorena M Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - James Lemon
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nicole Ingram
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ty W Turner
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ralf Dechend
- Experimental and Clinical Research Center, HELIOS Clinic, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center, HELIOS Clinic, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
9
|
Amruta N, Kandikattu HK, Intapad S. Cardiovascular Dysfunction in Intrauterine Growth Restriction. Curr Hypertens Rep 2022; 24:693-708. [PMID: 36322299 DOI: 10.1007/s11906-022-01228-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW We highlight important new findings on cardiovascular dysfunction in intrauterine growth restriction. RECENT FINDINGS Intrauterine growth restriction (IUGR) is a multifactorial condition which negatively impacts neonatal growth during pregnancy and is associated with health problems during the lifespan. It affects 5-15% of all pregnancies in the USA and Europe with varying percentages in developing countries. Epidemiological studies have reported that IUGR is associated with the pathogenesis of hypertension, activation of the renin-angiotensin system (RAS), disruption in placental-mTORC and TGFβ signaling cascades, and endothelial dysfunction in IUGR fetuses, children, adolescents, and adults resulting in the development of cardiovascular diseases (CVD). Experimental studies are needed to investigate therapeutic measures to treat increased blood pressure (BP) and long-term CVD problems in people affected by IUGR. We outline the mechanisms mediating fetal programming of hypertension in developing CVD. We have reviewed findings from different experimental models focusing on recent studies that demonstrate CVD in IUGR.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, #8683, New Orleans, LA, 70112-2699, USA
| | - Hemanth Kumar Kandikattu
- Department of Medicine, Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Suttira Intapad
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, #8683, New Orleans, LA, 70112-2699, USA.
| |
Collapse
|
10
|
Taylor EB, George EM. Animal Models of Preeclampsia: Mechanistic Insights and Promising Therapeutics. Endocrinology 2022; 163:6623845. [PMID: 35772781 PMCID: PMC9262036 DOI: 10.1210/endocr/bqac096] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 11/19/2022]
Abstract
Preeclampsia (PE) is a common pregnancy-specific disorder that is a major cause of both maternal and fetal morbidity and mortality. Central to the pathogenesis of PE is the production of antiangiogenic and inflammatory factors by the hypoxic placenta, leading to the downstream manifestations of the disease, including hypertension and end-organ damage. Currently, effective treatments are limited for PE; however, the development of preclinical animal models has helped in the development and evaluation of new therapeutics. In this review, we will summarize some of the more commonly used models of PE and highlight their similarities to the human syndrome, as well as the therapeutics tested in each model.
Collapse
Affiliation(s)
- Erin B Taylor
- Correspondence: Erin B. Taylor, PhD, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216-4505, USA.
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| |
Collapse
|
11
|
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
|
12
|
McClements L, Richards C, Patel N, Chen H, Sesperez K, Bubb KJ, Karlstaedt A, Aksentijevic D. Impact of reduced uterine perfusion pressure model of preeclampsia on metabolism of placenta, maternal and fetal hearts. Sci Rep 2022; 12:1111. [PMID: 35064159 PMCID: PMC8782944 DOI: 10.1038/s41598-022-05120-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/31/2021] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia is a cardiovascular pregnancy complication characterised by new onset hypertension and organ damage or intrauterine growth restriction. It is one of the leading causes of maternal and fetal mortality in pregnancy globally. Short of pre-term delivery of the fetus and placenta, treatment options are limited. Consequently, preeclampsia leads to increased cardiovascular disease risk in both mothers and offspring later in life. Here we aim to examine the impact of the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia on the maternal cardiovascular system, placental and fetal heart metabolism. The surgical RUPP model was induced in pregnant rats by applying silver clips around the aorta and uterine arteries on gestational day 14, resulting in ~ 40% uterine blood flow reduction. The experiment was terminated on gestational day 19 and metabolomic profile of placentae, maternal and fetal hearts analysed using high-resolution 1H NMR spectroscopy. Impairment of uterine perfusion in RUPP rats caused placental and cardiac hypoxia and a series of metabolic adaptations: altered energetics, carbohydrate, lipid and amino acid metabolism of placentae and maternal hearts. Comparatively, the fetal metabolic phenotype was mildly affected. Nevertheless, long-term effects of these changes in both mothers and the offspring should be investigated further in the future.
Collapse
Affiliation(s)
- Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Claire Richards
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Nikayla Patel
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hao Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kimberly Sesperez
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kristen J Bubb
- Biomedical Discovery Institute, Monash University, Melbourne, Australia
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA, 127 San Vincente Blvd, 90048
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
13
|
Wat JM, Baczyk D, Kingdom JC. The antithrombin binding regions of heparin mediate fetal growth and reduced placental damage in the RUPP model of preeclampsia†. Biol Reprod 2021; 102:1102-1110. [PMID: 31950133 PMCID: PMC7186778 DOI: 10.1093/biolre/ioaa006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
Preeclampsia is a serious hypertensive disorder of pregnancy, which is only cured with delivery of the placenta, thereby commonly necessitating preterm birth of the fetus. Low-molecular-weight heparin (LMWH) has demonstrated potential to reduce the incidence of preeclampsia in high-risk pregnant women, although the underlying mechanism by which LMWH protects against preeclampsia is unknown. Given the complex structure and biologic actions of heparin, we tested the hypothesis that heparin can mediate preeclampsia prevention via nonanticoagulant pathways. We compared the effects of a nonanticoagulant, glycol-split LMWH (gsHep)—rendered nonanticoagulant through disruption of the antithrombin binding regions—with the LMWH dalteparin in the rat reduced uterine perfusion pressure (RUPP) surgical model of preeclampsia. Although RUPP animals exhibit significantly elevated blood pressure and reduced plasma levels of placental growth factor (PGF) compared to sham, neither dalteparin nor gsHep treatment significantly impacted these parameters. However, the observed positive correlation between PGF levels and number of viable fetuses in RUPP-induced animals suggests that reduced PGF levels were predominately due to placental loss. Daily subcutaneous injections of low-dose dalteparin but not gsHep significantly restored fetal growth that was impaired by RUPP surgery. Placentas from RUPP animals exhibited an abnormal labyrinth structure, characterized by expanded sinusoidal blood spaces, relative to sham-operated animals. Morphometric analysis demonstrated that dalteparin but not gsHep treatment normalized development of the labyrinth in RUPP-exposed conceptuses. These data suggest that the antithrombin-binding regions of LMWH are required to confer its protective effects on fetal growth and placental development.
Collapse
Affiliation(s)
- Jovian M Wat
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dora Baczyk
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - John C Kingdom
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Maternal-Fetal Medicine Division, Department of Obstetrics and Gynaecology, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Richards C, Sesperez K, Chhor M, Ghorbanpour S, Rennie C, Ming CLC, Evenhuis C, Nikolic V, Orlic NK, Mikovic Z, Stefanovic M, Cakic Z, McGrath K, Gentile C, Bubb K, McClements L. Characterisation of cardiac health in the reduced uterine perfusion pressure model and a 3D cardiac spheroid model, of preeclampsia. Biol Sex Differ 2021; 12:31. [PMID: 33879252 PMCID: PMC8056582 DOI: 10.1186/s13293-021-00376-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Background Preeclampsia is a dangerous cardiovascular disorder of pregnancy that leads to an increased risk of future cardiovascular and metabolic disorders. Much of the pathogenesis and mechanisms involved in cardiac health in preeclampsia are unknown. A novel anti-angiogenic protein, FKBPL, is emerging as having a potential role in both preeclampsia and cardiovascular disease (CVD). Therefore, in this study we aimed to characterise cardiac health and FKBPL regulation in the rat reduced uterine perfusion pressure (RUPP) and a 3D cardiac spheroid model of preeclampsia. Methods The RUPP model was induced in pregnant rats and histological analysis performed on the heart, kidney, liver and placenta (n ≥ 6). Picrosirius red staining was performed to quantify collagen I and III deposition in rat hearts, placentae and livers as an indicator of fibrosis. RT-qPCR was used to determine changes in Fkbpl, Icam1, Vcam1, Flt1 and Vegfa mRNA in hearts and/or placentae and ELISA to evaluate cardiac brain natriuretic peptide (BNP45) and FKBPL secretion. Immunofluorescent staining was also conducted to analyse the expression of cardiac FKBPL. Cardiac spheroids were generated using human cardiac fibroblasts and human coronary artery endothelial cells and treated with patient plasma from normotensive controls, early-onset preeclampsia (EOPE) and late-onset preeclampsia (LOPE); n = 3. FKBPL and CD31 expression was quantified by immunofluorescent labelling. Results The RUPP procedure induced significant increases in blood pressure (p < 0.001), collagen deposition (p < 0.001) and cardiac BNP45 (p < 0.05). It also induced a significant increase in cardiac FKBPL mRNA (p < 0.05) and protein expression (p < 0.01). RUPP placentae also exhibited increased collagen deposition and decreased Flt1 mRNA expression (p < 0.05). RUPP kidneys revealed an increase in average glomerular size (p < 0.05). Cardiac spheroids showed a significant increase in FKBPL expression when treated with LOPE plasma (p < 0.05) and a trend towards increased FKBPL expression following treatment with EOPE plasma (p = 0.06). Conclusions The rat RUPP model induced cardiac, renal and placental features reflective of preeclampsia. FKBPL was increased in the hearts of RUPP rats and cardiac spheroids treated with plasma from women with preeclampsia, perhaps reflective of restricted angiogenesis and inflammation in this disorder. Elucidation of these novel FKBPL mechanisms in cardiac health in preeclampsia could be key in preventing future CVD. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-021-00376-1.
Collapse
Affiliation(s)
- Claire Richards
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kimberly Sesperez
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Michael Chhor
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Sahar Ghorbanpour
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Clara Liu Chung Ming
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Chris Evenhuis
- The iThree Institute, University of Technology Sydney, Sydney, NSW, Australia
| | - Valentina Nikolic
- Department of Pharmacology and Toxicology & Department of Internal Medicine - Gynaecology, Medical Faculty, University of Nis, Nis, Serbia
| | - Natasa Karadzov Orlic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia.,Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Zeljko Mikovic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia.,Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Milan Stefanovic
- Department of Pharmacology and Toxicology & Department of Internal Medicine - Gynaecology, Medical Faculty, University of Nis, Nis, Serbia.,Department of Gynaecology and Obstetrics, Clinical Centre Nis, Nis, Serbia
| | - Zoran Cakic
- Department of Gynaecology and Obstetrics, General Hospital of Leskovac, Leskovac, Serbia
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia.,The Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Kristen Bubb
- The Kolling Institute, University of Sydney, Sydney, NSW, Australia.,Biomedical Discovery Institute, Monash University, Melbourne, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Siragher E, Sferruzzi-Perri AN. Placental hypoxia: What have we learnt from small animal models? Placenta 2021; 113:29-47. [PMID: 34074553 DOI: 10.1016/j.placenta.2021.03.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 12/31/2022]
Abstract
Intrauterine hypoxia is a feature of pregnancy complications, both at high altitude and sea level. To understand the placental response to reduced oxygen availability, small animal models of maternal inhalation hypoxia (MIH) or reduced uterine perfusion pressure (RUPP) may be utilised. The aim of this review was to compare the findings of those studies to identify the role of oxygen availability in adapting placental structural and functional phenotypes in relation to fetal outcome. It also sought to explore the evidence for the involvement of particular genes and protein signalling pathways in the placenta in mediating hypoxia driven alterations. The data available demonstrate that both MIH and RUPP can induce placental hypoxia, which affects placental structure and vascularity, as well as glucose, amino acid, calcium and possibly lipid transport capacity. In addition, changes have been observed in HIF, VEGF, insulin/IGF2, AMPK, mTOR, PI3K and PPARγ signalling, which may be key in linking together observed phenotypes under conditions of placental hypoxia. Many different manipulations have been examined, with varied outcomes depending on the intensity, timing and duration of the insult. Some manipulations have detrimental effects on placental phenotype, viability and fetal growth, whereas in others, the placenta appears to adapt to uphold fetal growth despite the challenge of low oxygen. Together these data suggest a complex response of the placenta to reduced oxygen availability, which links to changes in fetal outcomes. However, further work is required to explore the role of fetal sex, altered maternal physiology and placental molecular mechanisms to fully understand placental responses to hypoxia and their relevance for pregnancy outcome.
Collapse
Affiliation(s)
- Emma Siragher
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
16
|
Mata K, Nobre AVV, Felix Silva PH, Oliezer RS, Fernandes C, Amaral J, Ramos J, Constante Gabriel Del-Arco M, Messora MR, Tanus-Santos JE, Gerlach RF, Salvador SL. A new mixed model of periodontitis-induced preeclampsia: A pilot study. J Periodontal Res 2021; 56:726-734. [PMID: 33686671 DOI: 10.1111/jre.12869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES/BACKGROUND Recent studies have shown that periodontal disease is strongly related to gestational complications such as preeclampsia (PE). PE is responsible for 42% of maternal deaths worldwide and kills approximately 76 000 women a year. In addition, children born under PE conditions are at increased risk of hospitalization due to metabolic disorders, epilepsy, and other complications. Numerous reviews and clinical studies on PE have been published, but the mechanisms underlying the relationship between periodontal disease and PE and the way periodontopathogens alter vascular response in pregnant women remain unclear. METHODS This study aims to verify whether periodontal disease induces PE by using the association of two periodontitis (PD) models: ligature and oral Porphyromonas gingivalis (P. gingivalis) W83 inoculation in Wistar rats. At gestational day 5, the ligature was placed on each mandibular first molar, which was followed by daily oral P. gingivalis inoculation for 15 days. At gestational day 19, urine was collected, and invasive arterial pressure was measured. The animals were euthanized, and plasma and tissues were collected. RESULTS After 15 days of the association of ligature and P. gingivalis inoculation, the animals presented the characteristic symptoms of PE: altered blood pressure, proteinuria, and change in litter size (number of pups) and pup weight when compared to the control group (p < .005). The PE animals also presented greater bone porosity, trabecular separation, and reduced bone volume in the hemimandibles, as well as altered inflammatory response. The level of cytokine IL-6 was higher in the PE group than in the control group (p < .005). CONCLUSION The association of two PD models effectively induced PE. To our knowledge, this is the first study on the oral use of P. gingivalis for PE induction. Our results support the importance of PD as a possible cause for PE development, opening an important new avenue to study cause and consequence relationships in inflammation and PE due to exposure to periodontal infection.
Collapse
Affiliation(s)
- Karina Mata
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.,University of Rio Verde, UniRV, Formosa, GO, Brazil
| | - Atila Vinícius Vitor Nobre
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Pedro Henrique Felix Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Rene Seabra Oliezer
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Cleverson Fernandes
- University of Rio Verde, UniRV, Formosa, GO, Brazil.,Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jefferson Amaral
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Junia Ramos
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Marina Constante Gabriel Del-Arco
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Michel Reis Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - José Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Raquel Fernanda Gerlach
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sergio Luiz Salvador
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| |
Collapse
|
17
|
Coats LE, Bamrick-Fernandez DR, Ariatti AM, Bakrania BA, Rawls AZ, Ojeda NB, Alexander BT. Stimulation of soluble guanylate cyclase diminishes intrauterine growth restriction in a rat model of placental ischemia. Am J Physiol Regul Integr Comp Physiol 2020; 320:R149-R161. [PMID: 33175587 DOI: 10.1152/ajpregu.00234.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Placental ischemia in preeclampsia (PE) results in hypertension and intrauterine growth restriction (IUGR). Stimulation of soluble guanylate cyclase (sGC) reduces blood pressure in the clinically relevant reduced uterine perfusion pressure (RUPP) rat model of PE, implicating involvement in RUPP-induced hypertension. However, the contribution of sGC in the development of IUGR in PE is not known. Thus, this study demonstrated the efficacy of Riociguat, an sGC stimulator, in IUGR reversion in the RUPP rat model of PE, and tested the hypothesis that improvement in fetal weight occurs in association with improvement in placental perfusion, placental morphology, and placental nutrient transport protein expression. Sham or RUPP surgery was performed at gestational day 14 (G14) with administration of vehicle (Sham or RUPP) or the sGC stimulator (Riociguat, 10 mg/kg/day sc; sGC-treated) until G20. Fetal weight was reduced (P = 0.004) at G20 in RUPP but not in sGC-treated RUPP compared with Sham, the control group. At G20, uterine artery resistance index (UARI) was increased (P = 0.010) in RUPP, indicating poor placental perfusion; proportional junctional zone surface area was elevated (P = 0.035), indicating impaired placental development. These effects were ameliorated in sGC-treated RUPP. Placental protein expression of nutrient transporter heart fatty acid-binding protein (hFABP) was increased (P = 0.008) in RUPP but not in sGC-treated RUPP, suggesting a compensatory mechanism to maintain normal neurodevelopment. Yet, UARI (P < 0.001), proportional junctional zone surface area (P = 0.013), and placental hFABP protein expression (P = 0.008) were increased in sGC-treated Sham, suggesting a potential adverse effect of Riociguat. Collectively, these results suggest sGC contributes to IUGR in PE.
Collapse
Affiliation(s)
- Laura E Coats
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Allison M Ariatti
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bhavisha A Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Adam Z Rawls
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
18
|
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
|
19
|
Travis OK, White D, Baik C, Giachelli C, Thompson W, Stubbs C, Greer M, Lemon JP, Williams JM, Cornelius DC. Interleukin-17 signaling mediates cytolytic natural killer cell activation in response to placental ischemia. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1036-R1046. [PMID: 32320265 DOI: 10.1152/ajpregu.00285.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
T-helper (TH)17s, IL-17, and cytolytic natural killer cells (cNKs) are increased in preeclampsia and contribute to the hypertension, inflammation, and fetal growth restriction that occurs in response to placental ischemia in the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia. As IL-17 stimulates NK cytotoxicity in vitro, we tested the hypothesis that IL-17 inhibition in RUPP rats would decrease cNK activation as a mechanism to improve maternal and fetal outcomes. On gestation day (GD) 14, rats undergoing RUPP received a miniosmotic pump infusing IL-17RC (100 pg/day), a soluble IL-17 receptor (RUPP + IL-17RC). On GD19, mean arterial pressure (MAP) was measured in normal pregnant (NP), RUPP, and RUPP + IL-17RC rats (n = 10-12/group), animals were euthanized, and blood and tissues were collected for analysis. MAP was 30% higher in RUPP compared with NP (P < 0.0001) and was 12% lower in RUPP + IL-17RC (P = 0.0007 vs. RUPP). Placental cytolytic NK cells were 132% higher in RUPP than in NP (P = 0.04 vs. NP) and were normalized in RUPP + IL-17RC (P = 0.03 vs. RUPP). Placental levels of TNF-α, a cNK-secreted cytokine, and macrophage inflammatory protein-3α (MIP-3α), a cNK chemokine, were higher in RUPP vs. NP and lower after IL-17 blockade. Placental VEGF was lower in RUPP vs. NP and was normalized in RUPP + IL-17RC. In vitro cytolytic activity of RUPP placental NKs was higher compared with NP and was blunted in RUPP + IL-17RC NKs. Finally, both fetal weight and placental weight were lower in RUPP compared with NP, and were improved in RUPP + IL-17RC. These data identify IL-17 as a mediator of cNK activation in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Olivia K Travis
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dakota White
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cedar Baik
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Chelsea Giachelli
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Willie Thompson
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cassandra Stubbs
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mallory Greer
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - James P Lemon
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan Michael Williams
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
20
|
Gatford KL, Andraweera PH, Roberts CT, Care AS. Animal Models of Preeclampsia: Causes, Consequences, and Interventions. Hypertension 2020; 75:1363-1381. [PMID: 32248704 DOI: 10.1161/hypertensionaha.119.14598] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preeclampsia is a common pregnancy complication, affecting 2% to 8% of pregnancies worldwide, and is an important cause of both maternal and fetal morbidity and mortality. Importantly, although aspirin and calcium are able to prevent preeclampsia in some women, there is no cure apart from delivery of the placenta and fetus, often necessitating iatrogenic preterm birth. Preclinical models of preeclampsia are widely used to investigate the causes and consequences of preeclampsia and to evaluate safety and efficacy of potential preventative and therapeutic interventions. In this review, we provide a summary of the published preclinical models of preeclampsia that meet human diagnostic criteria, including the development of maternal hypertension, together with new-onset proteinuria, maternal organ dysfunction, and uteroplacental dysfunction. We then discuss evidence from preclinical models for multiple causal factors of preeclampsia, including those implicated in early-onset and late-onset preeclampsia. Next, we discuss the impact of exposure to a preeclampsia-like environment for later maternal and progeny health. The presence of long-term impairment, particularly cardiovascular outcomes, in mothers and progeny after an experimentally induced preeclampsia-like pregnancy, implies that later onset or reduced severity of preeclampsia will improve later maternal and progeny health. Finally, we summarize published intervention studies in preclinical models and identify gaps in knowledge that we consider should be targets for future research.
Collapse
Affiliation(s)
- Kathryn L Gatford
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Prabha H Andraweera
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Claire T Roberts
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| | - Alison S Care
- From the Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Australia
| |
Collapse
|
21
|
Morton JS, Levasseur J, Ganguly E, Quon A, Kirschenman R, Dyck JRB, Fraser GM, Davidge ST. Characterisation of the Selective Reduced Uteroplacental Perfusion (sRUPP) Model of Preeclampsia. Sci Rep 2019; 9:9565. [PMID: 31266978 PMCID: PMC6606748 DOI: 10.1038/s41598-019-45959-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/20/2019] [Indexed: 12/03/2022] Open
Abstract
Preeclampsia is a complication of pregnancy characterised by gestational hypertension, proteinuria and/or end organ disease. The reduced uteroplacental perfusion (RUPP) model, via partial occlusion of the lower abdominal aorta, mimics insufficient placental perfusion as a primary causal characteristic of preeclampsia. However, a major limitation of the RUPP model is that perfusion is reduced to the entire hindquarters of the rat resulting in hindlimb ischemia. We hypothesised that clipping the uterine and ovarian arteries in the selective (s)RUPP model would provoke signs of preeclampsia while avoiding systemic ischemia. Sham, RUPP or sRUPP procedures were performed in pregnant Sprague Dawley rats on gestational day (GD)14. On GD21 uterine blood flow was significantly reduced in both the RUPP and sRUPP models while aortic flow was reduced only in RUPP. Both models resulted in increased MAP, increased vascular oxidative stress (superoxide generation), increased pro-inflammatory (RANTES) and reduced pro-angiogenic (endoglin) mediators. Vascular compliance and constriction were unaltered in either RUPP or sRUPP groups. In summary, refinements to the RUPP model simultaneously maintain the characteristic phenotype of preeclampsia and avoid peripheral ischemia; providing a useful tool which may be used to increase our knowledge and bring us closer to a solution for women affected by preeclampsia.
Collapse
Affiliation(s)
- J S Morton
- Faculty of Medicine and Dentistry, Dept. of Ob/Gyn, University of Alberta, Edmonton, AB, T6G 2S2, Canada.,Women and Children's Health Research Institute, Edmonton, AB, T6G 2R3, Canada
| | - J Levasseur
- Faculty of Medicine and Dentistry, Dept. of Pediatrics, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - E Ganguly
- Faculty of Medicine and Dentistry, Dept. of Ob/Gyn, University of Alberta, Edmonton, AB, T6G 2S2, Canada.,Faculty of Medicine and Dentistry, Dept. of Physiology, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,Women and Children's Health Research Institute, Edmonton, AB, T6G 2R3, Canada
| | - A Quon
- Faculty of Medicine and Dentistry, Dept. of Ob/Gyn, University of Alberta, Edmonton, AB, T6G 2S2, Canada.,Women and Children's Health Research Institute, Edmonton, AB, T6G 2R3, Canada
| | - R Kirschenman
- Faculty of Medicine and Dentistry, Dept. of Ob/Gyn, University of Alberta, Edmonton, AB, T6G 2S2, Canada.,Women and Children's Health Research Institute, Edmonton, AB, T6G 2R3, Canada
| | - J R B Dyck
- Faculty of Medicine and Dentistry, Dept. of Pediatrics, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - G M Fraser
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - S T Davidge
- Faculty of Medicine and Dentistry, Dept. of Ob/Gyn, University of Alberta, Edmonton, AB, T6G 2S2, Canada. .,Faculty of Medicine and Dentistry, Dept. of Physiology, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Women and Children's Health Research Institute, Edmonton, AB, T6G 2R3, Canada.
| |
Collapse
|
22
|
Natale BV, Mehta P, Vu P, Schweitzer C, Gustin K, Kotadia R, Natale DRC. Reduced Uteroplacental Perfusion Pressure (RUPP) causes altered trophoblast differentiation and pericyte reduction in the mouse placenta labyrinth. Sci Rep 2018; 8:17162. [PMID: 30464252 PMCID: PMC6249310 DOI: 10.1038/s41598-018-35606-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
This study characterized the effect of the reduced utero-placental perfusion pressure (RUPP) model of placental insufficiency on placental morphology and trophoblast differentiation at mid-late gestation (E14.5). Altered trophoblast proliferation, reduced syncytiotrophoblast gene expression, increased numbers of sinusoidal trophoblast giant cells, decreased Vegfa and decreased pericyte presence in the labyrinth were observed in addition to changes in maternal blood spaces, the fetal capillary network and reduced fetal weight. Further, the junctional zone was characterized by reduced spongiotrophoblast and glycogen trophoblast with increased trophoblast giant cells. Increased Hif-1α and TGF-β-3 in vivo with supporting hypoxia studies in trophoblast stem (TS) cells in vitro, support hypoxia as a contributing factor to the RUPP placenta phenotype. Together, this study identifies altered cell populations within the placenta that may contribute to the phenotype, and thus support the use of RUPP in the mouse as a model of placenta insufficiency. As such, this model in the mouse provides a valuable tool for understanding the phenotypes resulting from genetic manipulation of isolated cell populations to further understand the etiology of placenta insufficiency and fetal growth restriction. Further this study identifies a novel relationship between placental insufficiency and pericyte depletion in the labyrinth layer.
Collapse
Affiliation(s)
- Bryony V Natale
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Prutha Mehta
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Priscilla Vu
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Christina Schweitzer
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Katarina Gustin
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ramie Kotadia
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - David R C Natale
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada.
- Department of Obstetrics and Gynecology in Reproductive Sciences, Faculty of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
23
|
Sandgren JA, Deng G, Linggonegoro DW, Scroggins SM, Perschbacher KJ, Nair AR, Nishimura TE, Zhang SY, Agbor LN, Wu J, Keen HL, Naber MC, Pearson NA, Zimmerman KA, Weiss RM, Bowdler NC, Usachev YM, Santillan DA, Potthoff MJ, Pierce GL, Gibson-Corley KN, Sigmund CD, Santillan MK, Grobe JL. Arginine vasopressin infusion is sufficient to model clinical features of preeclampsia in mice. JCI Insight 2018; 3:99403. [PMID: 30282823 DOI: 10.1172/jci.insight.99403] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 08/17/2018] [Indexed: 12/29/2022] Open
Abstract
Copeptin, a marker of arginine vasopressin (AVP) secretion, is elevated throughout human pregnancies complicated by preeclampsia (PE), and AVP infusion throughout gestation is sufficient to induce the major phenotypes of PE in mice. Thus, we hypothesized a role for AVP in the pathogenesis of PE. AVP infusion into pregnant C57BL/6J mice resulted in hypertension, renal glomerular endotheliosis, intrauterine growth restriction, decreased placental growth factor (PGF), altered placental morphology, placental oxidative stress, and placental gene expression consistent with human PE. Interestingly, these changes occurred despite a lack of placental hypoxia or elevations in placental fms-like tyrosine kinase-1 (FLT1). Coinfusion of AVP receptor antagonists and time-restricted infusion of AVP uncovered a mid-gestational role for the AVPR1A receptor in the observed renal pathologies, versus mid- and late-gestational roles for the AVPR2 receptor in the blood pressure and fetal phenotypes. These findings demonstrate that AVP is sufficient to initiate phenotypes of PE in the absence of placental hypoxia, and indicate that AVP may mechanistically (independently, and possibly synergistically with hypoxia) contribute to the development of clinical signs of PE in specific subtypes of human PE. Additionally, they identify divergent and gestational time-specific signaling mechanisms that mediate the development of PE phenotypes in response to AVP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Donna A Santillan
- Department of Obstetrics & Gynecology.,University of Iowa Hospitals & Clinics Center for Hypertension Research
| | - Matthew J Potthoff
- Department of Pharmacology.,University of Iowa Hospitals & Clinics Center for Hypertension Research.,François M. Abboud Cardiovascular Research Center.,Fraternal Order of Eagles' Diabetes Research Center, and.,Obesity Research & Education Initiative, University of Iowa, Iowa City, Iowa USA
| | - Gary L Pierce
- Department of Health & Human Physiology.,University of Iowa Hospitals & Clinics Center for Hypertension Research.,François M. Abboud Cardiovascular Research Center
| | - Katherine N Gibson-Corley
- Department of Pathology.,University of Iowa Hospitals & Clinics Center for Hypertension Research.,Fraternal Order of Eagles' Diabetes Research Center, and
| | - Curt D Sigmund
- Department of Pharmacology.,University of Iowa Hospitals & Clinics Center for Hypertension Research.,François M. Abboud Cardiovascular Research Center.,Fraternal Order of Eagles' Diabetes Research Center, and.,Obesity Research & Education Initiative, University of Iowa, Iowa City, Iowa USA
| | - Mark K Santillan
- Department of Obstetrics & Gynecology.,University of Iowa Hospitals & Clinics Center for Hypertension Research
| | - Justin L Grobe
- Department of Pharmacology.,University of Iowa Hospitals & Clinics Center for Hypertension Research.,François M. Abboud Cardiovascular Research Center.,Fraternal Order of Eagles' Diabetes Research Center, and.,Obesity Research & Education Initiative, University of Iowa, Iowa City, Iowa USA
| |
Collapse
|
24
|
Eddy AC, Bidwell GL, George EM. Pro-angiogenic therapeutics for preeclampsia. Biol Sex Differ 2018; 9:36. [PMID: 30144822 PMCID: PMC6109337 DOI: 10.1186/s13293-018-0195-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/30/2018] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia is a pregnancy-induced hypertensive disorder resulting from abnormal placentation, which causes factors such as sFlt-1 to be released into the maternal circulation. Though anti-hypertensive drugs and magnesium sulfate can be given in an effort to moderate symptoms, the syndrome is not well controlled. A hallmark characteristic of preeclampsia, especially early-onset preeclampsia, is angiogenic imbalance resulting from an inappropriately upregulated sFlt-1 acting as a decoy receptor binding vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), reducing their bioavailability. Administration of sFlt-1 leads to a preeclamptic phenotype, and several models of preeclampsia also have elevated levels of plasma sFlt-1, demonstrating its role in driving the progression of this disease. Treatment with either VEGF or PlGF has been effective in attenuating hypertension and proteinuria in multiple models of preeclampsia. VEGF, however, may have overdose toxicity risks that have not been observed in PlGF treatment, suggesting that PlGF is a potentially safer therapeutic option. This review discusses angiogenic balance as it relates to preeclampsia and the studies which have been performed in order to alleviate the imbalance driving the maternal syndrome.
Collapse
Affiliation(s)
- Adrian C Eddy
- Department of Physiology and Biophysics, 2500 N State St, Jackson, MS, 39216, USA
| | - Gene L Bidwell
- Department of Cell and Molecular Biology, 2500 N State St, Jackson, MS, 39216, USA.,Department of Neurology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | - Eric M George
- Department of Physiology and Biophysics, 2500 N State St, Jackson, MS, 39216, USA. .,Department of Cell and Molecular Biology, 2500 N State St, Jackson, MS, 39216, USA.
| |
Collapse
|
25
|
Marshall SA, Hannan NJ, Jelinic M, Nguyen TP, Girling JE, Parry LJ. Animal models of preeclampsia: translational failings and why. Am J Physiol Regul Integr Comp Physiol 2018; 314:R499-R508. [DOI: 10.1152/ajpregu.00355.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia affects up to 8% of pregnancies worldwide and is a leading cause of both maternal and fetal morbidity and mortality. Our current understanding of the cause(s) of preeclampsia is far from complete, and the lack of a single reliable animal model that recapitulates all aspects of the disease further confounds our understanding. This is partially due to the heterogeneous nature of the disease, coupled with our evolving understanding of its etiology. Nevertheless, animal models are still highly relevant and useful tools that help us better understand the pathophysiology of specific aspects of preeclampsia. This review summarizes the various types and characteristics of animal models used to study preeclampsia, highlighting particular features of these models relevant to clinical translation. This review points out the strengths and limitations of these models to illustrate the importance of using the appropriate model depending on the research question.
Collapse
Affiliation(s)
- Sarah A. Marshall
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Natalie J. Hannan
- The Translational Obstetrics Group, Mercy Hospital for Women, Department of Obstetrics and Gynaecology, The University of Melbourne, Victoria, Australia
| | - Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Thy P.H. Nguyen
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E. Girling
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, The University of Melbourne and Royal Women’s Hospital, Parkville, Victoria, Australia
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
26
|
Nejabati HR, Latifi Z, Ghasemnejad T, Fattahi A, Nouri M. Placental growth factor (PlGF) as an angiogenic/inflammatory switcher: lesson from early pregnancy losses. Gynecol Endocrinol 2017; 33:668-674. [PMID: 28447504 DOI: 10.1080/09513590.2017.1318375] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Placental growth factor (PlGF) is an angiogenic factor which belongs to vascular endothelial growth factor (VEGF) family. In addition to the angiogenic function of PlGF, in some conditions such as preeclampsia and early pregnancy losses, it can induce inflammatory reactions which could be accompanied with reduced angiogenesis. Hence, it is crucial to investigate inflammatory and angiogenic switching states and understand underlying mechanisms. PlGF is expressed in endometrium, placenta and trophoblast cells and is involved in maturation of uterine NK cells. Up-regulation of PlGF directs VEGF to VEGFR-2 and reinforces angiogenesis. However, when VEGF/VEGFR-2 signaling pathway is impaired, PlGF may shift to severe inflammation and cause tissue damages which could lead to early pregnancy losses. Downregulation of PlGF has also been reported in pregnancy complications. In this review, we discussed the role of PlGF in embryo implantation failure and early pregnancy loss and also possible mechanisms regarding the role of PlGF in angiogenic/inflammatory switching in early pregnancy losses. Furthermore, we summarized the effects of various compounds on PlGF expression and briefly talked about its therapeutic potential that may be an opportunity for prevention of pregnancy loss.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- a Women's Reproductive Health Research Center
- b Department of Clinical Biochemistry , Faculty of Medicine , and
| | - Zeinab Latifi
- b Department of Clinical Biochemistry , Faculty of Medicine , and
| | | | - Amir Fattahi
- a Women's Reproductive Health Research Center
- c Department of Reproductive Biology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mohammad Nouri
- c Department of Reproductive Biology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
27
|
Paauw ND, Joles JA, Spradley FT, Bakrania B, Zsengeller ZK, Franx A, Verhaar MC, Granger JP, Lely AT. Exposure to placental ischemia impairs postpartum maternal renal and cardiac function in rats. Am J Physiol Regul Integr Comp Physiol 2017; 312:R664-R670. [PMID: 28202440 DOI: 10.1152/ajpregu.00510.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/25/2017] [Accepted: 02/12/2017] [Indexed: 02/06/2023]
Abstract
Women with a history of preeclampsia (PE) have an increased risk to develop cardiovascular and renal diseases later in life, but the mechanisms underlying this effect are unknown. In rats, we assessed whether placental ischemia results in long-term effects on the maternal cardiovascular and renal systems using the reduced uterine perfusion pressure (RUPP) model for PE. Sprague-Dawley rats received either a Sham or RUPP operation at gestational day 14 The rats were followed for 8 wk after delivery (Sham n = 12, RUPP n = 21) at which time mean arterial pressure (MAP; conscious), 24-h albuminuria, glomerular filtration rate (GFR; transcutaneous, FITC-sinistrin), and cardiac function (Vevo 770 system) were assessed. Subsequently, all rats were euthanized for mesenteric artery vasorelaxation and histology of heart and kidney. At 8 wk after delivery, there was no difference in MAP and albuminuria. However, RUPP rats showed a significantly reduced GFR [2.61 ± 0.53 vs. 3.37 ± 0.74 ml/min; P = 0.01]. Ultrasound showed comparable cardiac structure, but RUPP rats had a lower left ventricular ejection fraction (62 ± 7 vs. 69 ± 10%; P = 0.04). Heart and kidney histology was not different between Sham or RUPP rats. Furthermore, there were no differences in endothelial-dependent or -independent vasorelaxation. We show that exposure to placental ischemia in rats is accompanied by functional disturbances in maternal renal and cardiac function 8 wk after a preeclamptic pregnancy. However, these changes were not dependent on differences in blood pressure, small artery vasorelaxation, or cardiac and renal structure at this time point postpartum.
Collapse
Affiliation(s)
- Nina D Paauw
- Department of Obstetrics, Wilhelmina Children's Hospital Birth Center, University Medical Center Utrecht, Utrecht, The Netherlands;
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank T Spradley
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Bhavisha Bakrania
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Zsuzsanna K Zsengeller
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Arie Franx
- Department of Obstetrics, Wilhelmina Children's Hospital Birth Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joey P Granger
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - A Titia Lely
- Department of Obstetrics, Wilhelmina Children's Hospital Birth Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
28
|
Zhang LW, Warrington JP. Magnesium Sulfate Prevents Placental Ischemia-Induced Increases in Brain Water Content and Cerebrospinal Fluid Cytokines in Pregnant Rats. Front Neurosci 2016; 10:561. [PMID: 28008305 PMCID: PMC5143678 DOI: 10.3389/fnins.2016.00561] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022] Open
Abstract
Magnesium sulfate (MgSO4) is the most widely used therapy in the clinic to prevent the progression of preeclampsia, a hypertensive disorder of pregnancy, to eclampsia. Eclampsia, manifested as unexplained seizures and/or coma during pregnancy or postpartum, accounts for ~13% of maternal deaths worldwide. While MgSO4 continues to be used in the clinic, the mechanisms by which it exerts its protective actions are not well understood. In this study, we tested the hypothesis that MgSO4 protects against placental ischemia-induced increases in brain water content and cerebrospinal fluid cytokines. To test this hypothesis, MgSO4 was administered via mini-osmotic pump (60 mg/day, i.p.) to pregnant and placental ischemic rats, induced by mechanical reduction of uterine perfusion pressure, from gestational day 14–19. This treatment regimen of MgSO4 led to therapeutic level of 2.8 ± 0.6 mmol/L Mg in plasma. MgSO4 had no effect on improving placental ischemia-induced changes in mean arterial pressure, number of live fetuses, or fetal and placental weight. Placental ischemia increased, while MgSO4 prevented the increase in water content in the anterior cerebrum. Cytokine and chemokine levels were measured in the cerebrospinal fluid using a multi-plex assay. Results demonstrate that cerebrospinal fluid, obtained via the cisterna magna, had reduced protein, albumin, interleukin (IL)-17A, IL-18, IL-2, eotaxin, fractalkine, interferon gamma, vascular endothelial growth factor (VEGF), and macrophage inflammatory protein (MIP)-2 following MgSO4 treatment. These data support the hypothesis that MgSO4 offers neuroprotection by preventing placental ischemia-induced cerebral edema and reducing levels of cytokines/chemokines in the cerebrospinal fluid.
Collapse
Affiliation(s)
- Linda W Zhang
- Department of Physiology and Biophysics, University of Mississippi Medical Center Jackson, MS, USA
| | - Junie P Warrington
- Department of Physiology and Biophysics, University of Mississippi Medical Center Jackson, MS, USA
| |
Collapse
|
29
|
Spradley FT, Tan AY, Joo WS, Daniels G, Kussie P, Karumanchi SA, Granger JP. Placental Growth Factor Administration Abolishes Placental Ischemia-Induced Hypertension. Hypertension 2016; 67:740-7. [PMID: 26831193 DOI: 10.1161/hypertensionaha.115.06783] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/10/2016] [Indexed: 12/17/2022]
Abstract
Preeclampsia is a pregnancy-specific disorder of new-onset hypertension. Unfortunately, the most effective treatment is early delivery of the fetus and placenta. Placental ischemia appears central to the pathogenesis of preeclampsia because placental ischemia/hypoxia induced in animals by reduced uterine perfusion pressure (RUPP) or in humans stimulates release of hypertensive placental factors into the maternal circulation. The anti-angiogenic factor soluble fms-like tyrosine kinase-1 (sFlt-1), which antagonizes and reduces bioavailable vascular endothelial growth factor and placental growth factor (PlGF), is elevated in RUPP rats and preeclampsia. Although PlGF and vascular endothelial growth factor are both natural ligands for sFlt-1, vascular endothelial growth factor also has high affinity to VEGFR2 (Flk-1) causing side effects like edema. PlGF is specific for sFlt-1. We tested the hypothesis that PlGF treatment reduces placental ischemia-induced hypertension by antagonizing sFlt-1 without adverse consequences to the mother or fetus. On gestational day 14, rats were randomized to 4 groups: normal pregnant or RUPP±infusion of recombinant human PlGF (180 μg/kg per day; AG31, a purified, recombinant human form of PlGF) for 5 days via intraperitoneal osmotic minipumps. On day 19, mean arterial blood pressure and plasma sFlt-1 were higher and glomerular filtration rate lower in RUPP than normal pregnant rats. Infusion of recombinant human PlGF abolished these changes seen with RUPP along with reducing oxidative stress. These data indicate that the increased sFlt-1 and reduced PlGF resulting from placental ischemia contribute to maternal hypertension. Our novel finding that recombinant human PlGF abolishes placental ischemia-induced hypertension, without major adverse consequences, suggests a strong therapeutic potential for this growth factor in preeclampsia.
Collapse
Affiliation(s)
- Frank T Spradley
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.).
| | - Adelene Y Tan
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.)
| | - Woo S Joo
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.)
| | - Garrett Daniels
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.)
| | - Paul Kussie
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.)
| | - S Ananth Karumanchi
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.)
| | - Joey P Granger
- From the Department of Surgery (F.T.S.), Department of Physiology and Biophysics (F.T.S., J.P.G.), and Cardiovascular-Renal Research Center (F.T.S., J.P.G.), The University of Mississippi Medical Center, Jackson; Aggamin Biologics, New York, NY (A.Y.T., W.S.J., G.D., P.K.); and Beth Israel Deaconess Medical Center, Department of Nephrology, Harvard Medical School, Boston, MA (S.A.K.).
| |
Collapse
|
30
|
Effects of selective reduced uterine perfusion pressure in pregnant rats. Placenta 2015; 36:1450-4. [DOI: 10.1016/j.placenta.2015.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 10/17/2015] [Accepted: 10/23/2015] [Indexed: 11/18/2022]
|
31
|
Spradley FT, Palei AC, Granger JP. Immune Mechanisms Linking Obesity and Preeclampsia. Biomolecules 2015; 5:3142-76. [PMID: 26569331 PMCID: PMC4693273 DOI: 10.3390/biom5043142] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/07/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia (PE) is characterized by hypertension occurring after the twentieth week of pregnancy. It is a significant contributor to maternal and perinatal morbidity and mortality in developing countries and its pervasiveness is increasing within developed countries including the USA. However, the mechanisms mediating the pathogenesis of this maternal disorder and its rising prevalence are far from clear. A major theory with strong experimental evidence is that placental ischemia, resulting from inappropriate remodeling and widening of the maternal spiral arteries, stimulates the release of soluble factors from the ischemic placenta causing maternal endothelial dysfunction and hypertension. Aberrant maternal immune responses and inflammation have been implicated in each of these stages in the cascade leading to PE. Regarding the increased prevalence of this disease, it is becoming increasingly evident from epidemiological data that obesity, which is a state of chronic inflammation in itself, increases the risk for PE. Although the specific mechanisms whereby obesity increases the rate of PE are unclear, there are strong candidates including activated macrophages and natural killer cells within the uterus and placenta and activation in the periphery of T helper cells producing cytokines including TNF-α, IL-6 and IL-17 and the anti-angiogenic factor sFlt-1 and B cells producing the agonistic autoantibodies to the angiotensin type 1 receptor (AT1-aa). This review will focus on the immune mechanisms that have been implicated in the pathogenesis of hypertension in PE with an emphasis on the potential importance of inflammatory factors in the increased risk of developing PE in obese pregnancies.
Collapse
Affiliation(s)
- Frank T Spradley
- Department of Physiology and Biophysics, Cardiovascular-Renal Research Center, Women's Health Research Center, The University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Ana C Palei
- Department of Physiology and Biophysics, Cardiovascular-Renal Research Center, Women's Health Research Center, The University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Joey P Granger
- Department of Physiology and Biophysics, Cardiovascular-Renal Research Center, Women's Health Research Center, The University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
32
|
Fraser GM, Morton JS, Schmidt SM, Bourque S, Davidge ST, Davenport MH, Steinback CD. Reduced uterine perfusion pressure decreases functional capillary density in skeletal muscle. Am J Physiol Heart Circ Physiol 2015; 309:H2002-7. [PMID: 26475590 DOI: 10.1152/ajpheart.00641.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/13/2015] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to examine the functional and structural capillary density in the reduced uterine perfusion pressure (RUPP) model, which when performed during pregnancy is an established animal model of preeclampsia. We hypothesized that the RUPP model would be associated with capillary rarefaction and impaired capillary perfusion, which would be more pronounced in the pregnant state. Female Sprague-Dawley rats (n = 32) were randomized to nonpregnancy (Nonpregnant) or breeding (Pregnant) at 12 wk of age and again to RUPP or SHAM surgeries on gestational day (GD) 14 (or equivalent age in nonpregnant rats). On GD 20 (or equivalent), capillary structure and perfusion of the extensor digitorum longus were imaged using digital intravital video microscopy. Functional videos were analyzed by a blinded observer to measure capillary density, expressed as capillaries per millimeter intersecting three staggered reference lines (200 μm). Flow was scored as the percentage of capillaries having 1) continuous, 2) intermittent, or 3) stopped flow. Total capillary density was not different between groups. There was a main effect of RUPP surgery resulting in decreased continuous flow vessels (P < 0.01) and increased stopped flow (P < 0.01), which was driven by differences between pregnant animals (Continuous flow: pregnant SHAM 80.1 ± 7.8% vs. pregnant RUPP 67.8 ± 11.2%, P < 0.05) (Stopped flow: pregnant SHAM 8.7 ± 3.2% vs. pregnant RUPP 17.9 ± 5.7%, P < 0.01). Our results demonstrate that the RUPP surgery is associated with a decrease in functional capillary density in skeletal muscle that is more pronounced in the pregnant state, which may contribute to the vascular pathophysiology observed in preeclampsia.
Collapse
Affiliation(s)
- Graham M Fraser
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jude S Morton
- Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sydney M Schmidt
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada; Program for Pregnancy and Postpartum Health, Physical Activity and Diabetes Laboratory, Faculty of Physical Education and Recreation, University of Alberta, Edmonton, Alberta, Canada; and
| | - Stephane Bourque
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sandra T Davidge
- Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Margie H Davenport
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada; Program for Pregnancy and Postpartum Health, Physical Activity and Diabetes Laboratory, Faculty of Physical Education and Recreation, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Craig D Steinback
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada; Program for Pregnancy and Postpartum Health, Physical Activity and Diabetes Laboratory, Faculty of Physical Education and Recreation, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
33
|
Szalai G, Romero R, Chaiworapongsa T, Xu Y, Wang B, Ahn H, Xu Z, Chiang PJ, Sundell B, Wang R, Jiang Y, Plazyo O, Olive M, Tarca AL, Dong Z, Qureshi F, Papp Z, Hassan SS, Hernandez-Andrade E, Than NG. Full-length human placental sFlt-1-e15a isoform induces distinct maternal phenotypes of preeclampsia in mice. PLoS One 2015; 10:e0119547. [PMID: 25860260 PMCID: PMC4393117 DOI: 10.1371/journal.pone.0119547] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/30/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Most anti-angiogenic preeclampsia models in rodents utilized the overexpression of a truncated soluble fms-like tyrosine kinase-1 (sFlt-1) not expressed in any species. Other limitations of mouse preeclampsia models included stressful blood pressure measurements and the lack of postpartum monitoring. We aimed to 1) develop a mouse model of preeclampsia by administering the most abundant human placental sFlt-1 isoform (hsFlt-1-e15a) in preeclampsia; 2) determine blood pressures in non-stressed conditions; and 3) develop a survival surgery that enables the collection of fetuses and placentas and postpartum (PP) monitoring. METHODS Pregnancy status of CD-1 mice was evaluated with high-frequency ultrasound on gestational days (GD) 6 and 7. Telemetry catheters were implanted in the carotid artery on GD7, and their positions were verified by ultrasound on GD13. Mice were injected through tail-vein with adenoviruses expressing hsFlt-1-e15a (n = 11) or green fluorescent protein (GFP; n = 9) on GD8/GD11. Placentas and pups were delivered by cesarean section on GD18 allowing PP monitoring. Urine samples were collected with cystocentesis on GD6/GD7, GD13, GD18, and PPD8, and albumin/creatinine ratios were determined. GFP and hsFlt-1-e15a expression profiles were determined by qRT-PCR. Aortic ring assays were performed to assess the effect of hsFlt-1-e15a on endothelia. RESULTS Ultrasound predicted pregnancy on GD7 in 97% of cases. Cesarean section survival rate was 100%. Mean arterial blood pressure was higher in hsFlt-1-e15a-treated than in GFP-treated mice (∆MAP = 13.2 mmHg, p = 0.00107; GD18). Focal glomerular changes were found in hsFlt-1-e15a -treated mice, which had higher urine albumin/creatinine ratios than controls (109.3 ± 51.7 μg/mg vs. 19.3 ± 5.6 μg/mg, p = 4.4 x 10(-2); GD18). Aortic ring assays showed a 46% lesser microvessel outgrowth in hsFlt-1-e15a-treated than in GFP-treated mice (p = 1.2 x 10(-2)). Placental and fetal weights did not differ between the groups. One mouse with liver disease developed early-onset preeclampsia-like symptoms with intrauterine growth restriction (IUGR). CONCLUSIONS A mouse model of late-onset preeclampsia was developed with the overexpression of hsFlt-1-e15a, verifying the in vivo pathologic effects of this primate-specific, predominant placental sFlt-1 isoform. HsFlt-1-e15a induced early-onset preeclampsia-like symptoms associated with IUGR in a mouse with a liver disease. Our findings support that hsFlt-1-e15a is central to the terminal pathway of preeclampsia, and it can induce the full spectrum of symptoms in this obstetrical syndrome.
Collapse
Affiliation(s)
- Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Bing Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Hyunyoung Ahn
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Po Jen Chiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Birgitta Sundell
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Rona Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Yang Jiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Olesya Plazyo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Mary Olive
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Computer Science, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Faisal Qureshi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
34
|
Murphy SR, Cockrell K. Regulation of soluble fms-like tyrosine kinase-1 production in response to placental ischemia/hypoxia: role of angiotensin II. Physiol Rep 2015; 3:3/2/e12310. [PMID: 25716926 PMCID: PMC4393214 DOI: 10.14814/phy2.12310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
While soluble fms-like tyrosine-1 (sFlt-1) is implicated in the pathogenesis of hypertension during preeclampsia, the mechanisms leading to the enhanced sFlt-1 production remain unclear. A recent report suggests exogenous angiotensin II (ANGII) stimulates sFlt-1 production in pregnant rats, however, the role of endogenous ANGII in mediating the placental production of sFlt-1 in response to placental ischemia remains unknown. Therefore, the purpose of this study was to determine the role of endogenous ANGII in mediating the placental production of sFlt-1 in response to placental ischemia in pregnant Sprague–Dawley rats. To this end we compared sFlt-1 and ANGII levels from placental explants collected from normal pregnant (NP) and Reduced Uterine Perfusion Pressure (RUPP) rats. sFlt-1 (3271 ± 264 vs. 2228 ± 324 pg/mL, P < 0.05) and ANGII levels (43.2 ± 2.8 vs. 26.7 ± 1.9 pg/mL, P < 0.05) were higher in placental explants from RUPP rats versus NP rats. Administration of Losartan, an angiotensin type 1 (AT1) receptor antagonist, (10 mg/day for 5 days) to RUPP rats significantly reduced plasma levels of sFlt-1 (1432 ± 255 pg/mL, P < 0.05) when compared with untreated control rats (3431 ± 454 pg/mL). In addition, RUPP-induced hypertension was significantly reduced (113 ± 2 mmHg vs. 139 ± 2 mmHg, P < 0.05). In conclusion, placental sFlt-1 and ANGII production are significantly elevated in response to placental ischemia in pregnant rats. In addition, AT1 receptor activation, by endogenous ANGII, appears to play an important role in mediating the placental production of sFlt-1 in response to placental ischemia in pregnant rats.
Collapse
Affiliation(s)
- Sydney R Murphy
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kathy Cockrell
- Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
35
|
Magnesium sulfate treatment reverses seizure susceptibility and decreases neuroinflammation in a rat model of severe preeclampsia. PLoS One 2014; 9:e113670. [PMID: 25409522 PMCID: PMC4237502 DOI: 10.1371/journal.pone.0113670] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/27/2014] [Indexed: 12/20/2022] Open
Abstract
Eclampsia, defined as unexplained seizure in a woman with preeclampsia, is a life-threatening complication of pregnancy with unclear etiology. Magnesium sulfate (MgSO4) is the leading eclamptic seizure prophylactic, yet its mechanism of action remains unclear. Here, we hypothesized severe preeclampsia is a state of increased seizure susceptibility due to blood-brain barrier (BBB) disruption and neuroinflammation that lowers seizure threshold. Further, MgSO4 decreases seizure susceptibility by protecting the BBB and preventing neuroinflammation. To model severe preeclampsia, placental ischemia (reduced uteroplacental perfusion pressure; RUPP) was combined with a high cholesterol diet (HC) to cause maternal endothelial dysfunction. RUPP+HC rats developed symptoms associated with severe preeclampsia, including hypertension, oxidative stress, endothelial dysfunction and fetal and placental growth restriction. Seizure threshold was determined by quantifying the amount of pentylenetetrazole (PTZ; mg/kg) required to elicit seizure in RUPP+HC±MgSO4 and compared to normal pregnant controls (n = 6/group; gestational day 20). RUPP+HC rats were more sensitive to PTZ with seizure threshold being ∼65% lower vs. control (12.4±1.7 vs. 36.7±3.9 mg/kg PTZ; p<0.05) that was reversed by MgSO4 (45.7±8.7 mg/kg PTZ; p<0.05 vs. RUPP+HC). BBB permeability to sodium fluorescein, measured in-vivo (n = 5–7/group), was increased in RUPP+HC vs. control rats, with more tracer passing into the brain (15.9±1.0 vs. 12.2±0.3 counts/gram ×1000; p<0.05) and was unaffected by MgSO4 (15.6±1.0 counts/gram ×1000; p<0.05 vs. controls). In addition, RUPP+HC rats were in a state of neuroinflammation, indicated by 35±2% of microglia being active compared to 9±2% in normal pregnancy (p<0.01; n = 3–8/group). MgSO4 treatment reversed neuroinflammation, reducing microglial activation to 6±2% (p<0.01 vs. RUPP+HC). Overall, RUPP+HC rats were in a state of augmented seizure susceptibility potentially due to increased BBB permeability and neuroinflammation. MgSO4 treatment reversed this, increasing seizure threshold and decreasing neuroinflammation, without affecting BBB permeability. Thus, reducing neuroinflammation may be one mechanism by which MgSO4 prevents eclampsia during severe preeclampsia.
Collapse
|
36
|
Chen M, Dasgupta C, Xiong F, Zhang L. Epigenetic upregulation of large-conductance Ca2+-activated K+ channel expression in uterine vascular adaptation to pregnancy. Hypertension 2014; 64:610-8. [PMID: 24914190 DOI: 10.1161/hypertensionaha.114.03407] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our previous study demonstrated that pregnancy increased large-conductance Ca(2+)-activated potassium channel β1 subunit (BKβ1) expression and large-conductance Ca(2+)-activated potassium channel activity in uterine arteries, which were abrogated by chronic hypoxia. The present study tested the hypothesis that promoter methylation/demethylation is a key mechanism in epigenetic reprogramming of BKβ1 expression patterns in uterine arteries. Ovine BKβ1 promoter of 2315 bp spanning from -2211 to +104 of the transcription start site was cloned, and an Sp1-380 binding site that contains CpG dinucleotide in its core binding sequences was identified. Site-directed deletion of the Sp1 site significantly decreased the BKβ1 promoter activity. Estrogen receptor-α bound to the Sp1 site through tethering to Sp1 and upregulated the expression of BKβ1. The Sp1 binding site at BKβ1 promoter was highly methylated in uterine arteries of nonpregnant sheep, and methylation inhibited transcription factor binding and BKβ1 promoter activity. Pregnancy caused a significant decrease in CpG methylation at the Sp1 binding site and increased Sp1 binding to the BKβ1 promoter and BKβ1 mRNA abundance. Chronic hypoxia during gestation abrogated this pregnancy-induced demethylation and upregulation of BKβ1 expression. The results provide evidence of a novel mechanism of promoter demethylation in pregnancy-induced reprogramming of large-conductance Ca(2+)-activated potassium channel expression and function in uterine arteries and suggest new insights of epigenetic mechanisms linking gestational hypoxia to aberrant uteroplacental circulation and increased risk of preeclampsia.
Collapse
Affiliation(s)
- Man Chen
- From the Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Chiranjib Dasgupta
- From the Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Fuxia Xiong
- From the Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Lubo Zhang
- From the Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, CA.
| |
Collapse
|
37
|
Conrad KP, Davison JM. The renal circulation in normal pregnancy and preeclampsia: is there a place for relaxin? Am J Physiol Renal Physiol 2014; 306:F1121-35. [PMID: 24647709 DOI: 10.1152/ajprenal.00042.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
During the first trimester of human pregnancy, the maternal systemic circulation undergoes remarkable vasodilation. The kidneys participate in this vasodilatory response resulting in marked increases in renal plasma flow (RPF) and glomerular filtration rate (GFR). Comparable circulatory adaptations are observed in conscious gravid rats. Administration of the corpus luteal hormone relaxin (RLN) to nonpregnant rats and humans elicits vasodilatory changes like those of pregnancy. Systemic and renal vasodilation are compromised in midterm pregnant rats by neutralization or elimination of circulating RLN and in women conceiving with donor eggs who lack a corpus luteum and circulating RLN. Although RLN exerts both rapid (minutes) and sustained (hours to days) vasodilatory actions through different molecular mechanisms, a final common pathway is endothelial nitric oxide. In preeclampsia (PE), maternal systemic and renal vasoconstriction leads to hypertension and modest reduction in GFR exceeding that of RPF. Elevated level of circulating soluble vascular endothelial growth factor receptor-1 arising from the placenta is implicated in the hypertension and disruption of glomerular fenestrae and barrier function, the former causing reduced Kf and the latter proteinuria. Additional pathogenic factors are discussed. Last, potential clinical ramifications include RLN replacement in women conceiving with donor eggs and its therapeutic use in PE. Another goal has been to apply knowledge gained from investigating circulatory adaptations in pregnancy toward identifying and developing novel therapeutic strategies for renal and cardiovascular disease in the nonpregnant population. So far, one candidate to emerge is RLN and its potential therapeutic use in heart failure.
Collapse
Affiliation(s)
- Kirk P Conrad
- Departments of Physiology and Functional Genomics and Obstetrics and Gynecology, D. H. Barron Reproductive and Perinatal Biology Research Program, University of Florida College of Medicine, Gainesville, Florida; and
| | - John M Davison
- Institute of Cellular Medicine and Royal Victoria Infirmary, Newcastle University and Newcastle Hospitals National Health Service Foundation Trust, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
38
|
George EM, Granger JP. Recent insights into the pathophysiology of preeclampsia. ACTA ACUST UNITED AC 2014; 5:557-566. [PMID: 21170149 DOI: 10.1586/eog.10.45] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Preeclampsia, characterized by new-onset gestational hypertension and proteinuria, is a common and serious complication of pregnancy. Evidence from both animal and human studies has implicated placental ischemia and hypoxia as a central causative factor in the etiology of the disorder. The ischemic placenta in turn initiates a cascade of secondary effector mechanisms, including altered proangiogenic and antiangiogenic factor balance, increase in maternal oxidative stress and endothelial and immunological dysfunction. The full elucidation of these mechanisms will hopefully lead to a more complete understanding of the etiology of preeclampsia and lead to successful therapeutic intervention through the targeted disruption of new and novel pathways.
Collapse
Affiliation(s)
- Eric M George
- Department of Physiology and Biophysics and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | | |
Collapse
|
39
|
Amaral LM, Pinheiro LC, Guimaraes DA, Palei ACT, Sertório JT, Portella RL, Tanus-Santos JE. Antihypertensive effects of inducible nitric oxide synthase inhibition in experimental pre-eclampsia. J Cell Mol Med 2013; 17:1300-7. [PMID: 23890248 PMCID: PMC4159028 DOI: 10.1111/jcmm.12106] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/27/2013] [Indexed: 01/06/2023] Open
Abstract
Upregulation of inducible nitric oxide synthase (iNOS) has been reported in both experimental and clinical hypertension. However, although pro-inflammatory cytokines that up-regulate iNOS contribute to pre-eclampsia, no previous study has tested the hypothesis that a selective iNOS inhibitor (1400 W) could exert antihypertensive effects associated with decreased iNOS expression and nitrosative stress in pre-eclampsia. This study examined the effects of 1400 W in the reduced uteroplacental perfusion pressure (RUPP) placental ischaemia animal model and in normal pregnant rats. Sham-operated and RUPP rats were treated with daily vehicle or 1 mg/kg/day N-[3-(Aminomethyl) benzyl] acetamidine (1400 W) subcutaneously for 5 days. Plasma 8-isoprostane levels, aortic reactive oxygen species (ROS) levels and nicotinamide adenine dinucleotide phosphate (NADPH)-dependent ROS production were evaluated by ELISA, dihydroethidium fluorescence microscopy and lucigenin chemiluminescence respectively. Inducible nitric oxide synthase expression was assessed by western blotting analysis and aortic nitrotyrosine was evaluated by immunohistochemistry. Mean arterial blood pressure increased by ∼30 mmHg in RUPP rats, and 1400 W attenuated this increase by ∼50% (P < 0.05). While RUPP increased plasma 8-isoprostane levels, aortic ROS levels, and NADPH-dependent ROS production (P < 0.05), treatment with 1400 W blunted these alterations (P < 0.05). Moreover, while RUPP increased iNOS expression and aortic nitrotyrosine levels (P < 0.05), treatment with 1400 W blunted these alterations (P < 0.05). These results clearly implicate iNOS in the hypertension associated with RUPP. Our findings may suggest that iNOS inhibitors could be clinically useful in the therapy of pre-eclampsia, especially in particular groups of patients genetically more prone to express higher levels of iNOS. This issue deserves further confirmation.
Collapse
Affiliation(s)
- Lorena M Amaral
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
40
|
George EM, Palei AC, Dent EA, Granger JP. Sildenafil attenuates placental ischemia-induced hypertension. Am J Physiol Regul Integr Comp Physiol 2013; 305:R397-403. [PMID: 23785075 DOI: 10.1152/ajpregu.00216.2013] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia is a complication of pregnancy that is marked by hypertension, proteinuria, and maternal endothelial dysfunction. A central factor in the etiology of the disease is the development of placental hypoxia/ischemia, which releases pathogenic soluble factors. There is currently no effective treatment for preeclampsia, but the phosphodiesterase-5 (PDE-5) inhibitor sildenafil has been suggested, as PDE-5 is enriched in the uterus, and its antagonism could improve uteroplacental function. Here, we report in the reduced uterine perfusion pressure (RUPP) rat model that administration of oral sildenafil is effective in attenuating placental ischemia-induced hypertension during gestation. RUPP animals have significantly elevated arterial pressure compared with control animals (132 ± 3 vs. 100 ± 2 mmHg; P < 0.05). Administration of oral sildenafil (45 mg·kg⁻¹·day⁻¹) had no effect on blood pressure in control rats but decreased pressure in RUPP rats (115 ± 1 mmHg; P < 0.05). RUPP induced changes in placental sFlt-1, and vascular endothelial growth factor (VEGF) was unaffected by sildenafil administration, as was the decrease in free plasma VEGF. RUPP animals had a significant increase in medullary PDE-5/β-actin ratio (1 ± 0.14 vs. 1.63 ± 0.18; P < 0.05) expression with a resulting reduction in renal medullary cGMP (1.5 ± 0.15 vs. 0.99 ± 0.1 pmol/μg protein, P < 0.05) compared with controls. Although sildenafil had no effect on renal medullary cGMP in control animals, it significantly increased cGMP in RUPP animals (1.3 ± 0.1 pmol/μg protein; P < 0.05). These data suggest that sildenafil might provide an effective therapeutic option for the management of hypertension during preeclampsia.
Collapse
Affiliation(s)
- Eric M George
- Department of Physiology and the Cardiovascular Renal Research Center, The University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | | |
Collapse
|
41
|
Murphy SR, LaMarca BBD, Parrish M, Cockrell K, Granger JP. Control of soluble fms-like tyrosine-1 (sFlt-1) production response to placental ischemia/hypoxia: role of tumor necrosis factor-α. Am J Physiol Regul Integr Comp Physiol 2012. [PMID: 23193111 DOI: 10.1152/ajpregu.00069.2012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although abnormal soluble fms-like tyrosine kinase-1 (sFlt-1) production is thought to be an important factor in the pathogenesis of preeclampsia (PE), the mechanisms that regulate the production of sFlt-1 during PE are unclear. While our laboratory has shown tumor necrosis factor-α (TNF-α) and sFlt-1 to be elevated in pregnant rats in response to placental ischemia, the importance of TNF-α in the regulation of sFlt-1 production is unknown. Therefore, the purpose of this study was to determine the role of TNF-α in mediating the increase in sFlt-1 in response to placental ischemia or hypoxia. Reductions in uterine perfusion pressure in pregnant rats significantly increased plasma levels of sFlt-1 and tended to increase TNF-α, an effect markedly attenuated by pretreatment with a TNF-α inhibitor etanercept (0.4 mg/kg). To further assess chronic interactions between TNF-α and sFlt-1, we examined a chronic effect of TNF-α infusion (50 ng/day) into normal pregnant rats to increase plasma sFlt-1 levels, as well as the effects of acute hypoxia on placental sFlt-1 production in the absence and presence of TNF-α blockade. Placental explants exposed to hypoxic conditions had enhanced TNF-α levels versus normoxic conditions, as well as increased sFlt-1 production. Pretreatment of placental explants with etanercept (15 μM) significantly reduced TNF-α levels in response to hypoxia but did not attenuate sFlt-1 production. These data suggest that while TNF-α may not play an important role in stimulating sFlt-1 production in response to acute hypoxia, a more chronic hypoxia, or placental ischemia may be an important stimulus for enhanced sFlt-l production.
Collapse
Affiliation(s)
- Sydney R Murphy
- Dept. of Physiology and Biophysics, Univ. of Mississippi Medical Center, Jackson, MS 39216-4505, USA
| | | | | | | | | |
Collapse
|
42
|
Dasgupta C, Chen M, Zhang H, Yang S, Zhang L. Chronic hypoxia during gestation causes epigenetic repression of the estrogen receptor-α gene in ovine uterine arteries via heightened promoter methylation. Hypertension 2012; 60:697-704. [PMID: 22777938 DOI: 10.1161/hypertensionaha.112.198242] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Estrogen receptor-α (ERα) plays a key role in the adaptation of increased uterine blood flow in pregnancy. Chronic hypoxia is a common stress to maternal cardiovascular homeostasis and causes increased risk of preeclampsia. Studies in pregnant sheep demonstrated that hypoxia during gestation downregulated ERα gene expression in uterine arteries. The present study tested the hypothesis that hypoxia causes epigenetic repression of the ERα gene in uterine arteries via heightened promoter methylation. Ovine ERα promoter of 2035 bp spanning from -2000 to +35 of the transcription start site was cloned. No estrogen or hypoxia-inducible factor response elements were found at the promoter. Two transcription factor binding sites, USF(-15) and Sp1(-520), containing CpG dinucleotides were identified, which had significant effects on the promoter activity. The USF element binds transcription factors USF1 and USF2, and the Sp1 element binds Sp1, as well as ERα through Sp1. Deletion of the Sp1 site abrogated 17β-estradiol-induced increase in the promoter activity. In normoxic control sheep, CpG methylation at the Sp1 but not the USF site was significantly decreased in uterine arteries of pregnant as compared with nonpregnant animals. In pregnant sheep exposed to long-term high-altitude hypoxia, CpG methylation at both Sp1 and USF sites in uterine arteries was significantly increased. Methylation inhibited transcription factor binding and the promoter activity. The results provide evidence of hypoxia causing heightened promoter methylation and resultant ERα gene repression in uterine arteries and suggest new insights of molecular mechanisms linking gestational hypoxia to aberrant uteroplacental circulation and increased risk of preeclampsia.
Collapse
Affiliation(s)
- Chiranjib Dasgupta
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | |
Collapse
|
43
|
Hu XQ, Xiao D, Zhu R, Huang X, Yang S, Wilson SM, Zhang L. Chronic hypoxia suppresses pregnancy-induced upregulation of large-conductance Ca2+-activated K+ channel activity in uterine arteries. Hypertension 2012; 60:214-22. [PMID: 22665123 DOI: 10.1161/hypertensionaha.112.196097] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Our previous study demonstrated that increased Ca(2+)-activated K(+) (BK(Ca)) channel activity played a key role in the normal adaptation of reduced myogenic tone of uterine arteries in pregnancy. The present study tested the hypothesis that chronic hypoxia during gestation inhibits pregnancy-induced upregulation of BK(Ca) channel function in uterine arteries. Resistance-sized uterine arteries were isolated from nonpregnant and near-term pregnant sheep maintained at sea level (≈ 300 m) or exposed to high-altitude (3801 m) hypoxia for 110 days. Hypoxia during gestation significantly inhibited pregnancy-induced upregulation of BK(Ca) channel activity and suppressed BK(Ca) channel current density in pregnant uterine arteries. This was mediated by a selective downregulation of BK(Ca) channel β1 subunit in the uterine arteries. In accordance, hypoxia abrogated the role of the BK(Ca) channel in regulating pressure-induced myogenic tone of uterine arteries that was significantly elevated in pregnant animals acclimatized to chronic hypoxia. In addition, hypoxia abolished the steroid hormone-mediated increase in the β1 subunit and BK(Ca) channel current density observed in nonpregnant uterine arteries. Although the activation of protein kinase C inhibited BK(Ca) channel current density in pregnant uterine arteries of normoxic sheep, this effect was ablated in the hypoxic animals. The results demonstrate that selectively targeting BK(Ca) channel β1 subunit plays a critical role in the maladaption of uteroplacental circulation caused by chronic hypoxia, which contributes to the increased incidence of preeclampsia and fetal intrauterine growth restriction associated with gestational hypoxia.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Lectin-Like Oxidized Low-Density Lipoprotein 1 Receptor in a Reduced Uteroplacental Perfusion Pressure Rat Model of Preeclampsia. Hypertension 2012; 59:1014-20. [DOI: 10.1161/hypertensionaha.112.191825] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
45
|
Li J, LaMarca B, Reckelhoff JF. A model of preeclampsia in rats: the reduced uterine perfusion pressure (RUPP) model. Am J Physiol Heart Circ Physiol 2012; 303:H1-8. [PMID: 22523250 DOI: 10.1152/ajpheart.00117.2012] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Preeclampsia is defined as new-onset hypertension with proteinuria after 20 wk gestation and is hypothesized to be due to shallow trophoblast invasion in the spiral arteries thus resulting in progressive placental ischemia as the fetus grows. Many animal models have been developed that mimic changes in maternal circulation or immune function associated with preeclampsia. The model of reduced uterine perfusion pressure in pregnant rats closely mimics the hypertension, immune system abnormalities, systemic and renal vasoconstriction, and oxidative stress in the mother, and intrauterine growth restriction found in the offspring. The model has been successfully used in many species; however, rat and primate are the most consistent in comparison of characteristics with human preeclampsia. The model suffers, however, from lack of the ability to study the mechanisms responsible for abnormal placentation that ultimately leads to placental ischemia. Despite this limitation, the model is excellent for studying the consequences of reduced uterine blood flow as it mimics many of the salient features of preeclampsia during the last weeks of gestation in humans. This review discusses these features.
Collapse
Affiliation(s)
- Jing Li
- Women's Health Research Center, Jackson, Mississippi, USA
| | | | | |
Collapse
|
46
|
George EM, Arany I. Induction of heme oxygenase-1 shifts the balance from proinjury to prosurvival in the placentas of pregnant rats with reduced uterine perfusion pressure. Am J Physiol Regul Integr Comp Physiol 2012; 302:R620-6. [PMID: 22237591 DOI: 10.1152/ajpregu.00617.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Placental hypoxia/ischemia has been implicated as a central factor in the development of preeclampsia. One particularly useful animal model to study the impact of placental ischemia is the reduced uterine perfusion pressure (RUPP) model. We have previously demonstrated that RUPP animals exhibit elevated placental oxidative stress, which plays an important role in the development of the associated maternal hypertension. Recently, we have demonstrated that cobalt protoporphyrin (CoPP)-mediated induction of heme oxygenase-1 (HO-1) attenuates RUPP-induced oxidative stress and consequent hypertension. However, signaling pathways that are involved in this process are virtually unknown. Here, we show that placentas from RUPP animals exhibit increased phosphorylation of JNK, STAT1, STAT3, and p52shc with a concomitant increase in caspase-3 activation and depletion of intracellular ATP. Treatment with CoPP decreased RUPP-induced phosphorylation of JNK and STAT1, while it increased phosphorylation of ERK and STAT3, leading to decreased caspase-3 activation and restoration of intracellular ATP content. Our data imply that RUPP induces oxidative stress and the consequent injurious state by increasing phosphorylation of mediators of injury (STAT1, JNK) and, to a lesser extent, survival (STAT3, p52shc) in placentas of pregnant rats. HO-1 induction shifts this balance to a prosurvival phenotype by augmenting phosphorylation of the prosurvival ERK and STAT3, while suppressing phosphorylation of JNK and STAT1. This attenuates the resulting injury, as indicated by caspase-3 activation and ATP depletion. These results demonstrate a novel therapeutic activity of HO-1 induction in placental cell survival during ischemia and support the HO-1 pathway as a promising therapeutic target for the management of preeclampsia.
Collapse
Affiliation(s)
- Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | |
Collapse
|
47
|
Reho JJ, Toot JD, Peck J, Novak J, Yun YH, Ramirez RJ. Increased Myogenic Reactivity of Uterine Arteries from Pregnant Rats with Reduced Uterine Perfusion Pressure. Pregnancy Hypertens 2011; 2:106-114. [PMID: 22679605 DOI: 10.1016/j.preghy.2011.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The etiology of preeclampsia remains unknown. However, a contributing factor to this hypertensive disease of pregnancy is a reduction in uterine perfusion pressure resulting in placental ischemia. Uterine arteries may be a major regulator of this process through changes in vascular reactivity and localized blood flow. The reduced uterine perfusion pressure (RUPP) pregnant rat is an established animal model of preeclampsia pathology. Pregnant Sprague Dawley rats were used for this investigation and subjected to RUPP or SHAM surgery on Day 14 of gestation. On Day 21 of gestation, animals were terminated and resistance-caliber uterine arteries were harvested and mounted on a pressurized arteriograph to examine myogenic reactivity, agonist induced vasodilation (methacholine and VEGF), and vasoconstriction (phenylephrine and U-46619). Resistance-caliber uterine arteries from RUPP animals exhibited increased myogenic reactivity and decreased vasodilation (methacholine and VEGF) compared to SHAM uterine arteries (p<0.05). Phenylephrine and U-46619 induced constriction was similar in uterine arteries between RUPP and SHAM rats. These results suggest that resistancecaliber uterine arteries from RUPP pregnant rats are altered to reflect a more constrictive phenotype which may play a role in the development of maternal hypertension demonstrated in these animals and thereby potentially in preeclampsia.
Collapse
Affiliation(s)
- John J Reho
- Program in Integrated Biosciences, The University of Akron, Akron, OH 44325
| | | | | | | | | | | |
Collapse
|
48
|
Ryan MJ, Gilbert EL, Glover PH, George EM, Masterson CW, McLemore GR, LaMarca B, Granger JP, Drummond HA. Placental ischemia impairs middle cerebral artery myogenic responses in the pregnant rat. Hypertension 2011; 58:1126-31. [PMID: 22068864 DOI: 10.1161/hypertensionaha.111.181453] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One potential mechanism contributing to the increased risk for encephalopathies in women with preeclampsia is altered cerebral vascular autoregulation resulting from impaired myogenic tone. Whether placental ischemia, a commonly proposed initiator of preeclampsia, alters cerebral vascular function is unknown. This study tested the hypothesis that placental ischemia in pregnant rats (caused by reduced uterine perfusion pressure [RUPP]) leads to impaired myogenic responses in middle cerebral arteries. Mean arterial pressure was increased by RUPP (135±3 mm Hg) compared with normal pregnant rats (103±2 mm Hg) and nonpregnant controls (116±1 mm Hg). Middle cerebral arteries from rats euthanized on gestation day 19 were assessed in a pressure arteriograph under active (+Ca(2+)) and passive (0 Ca(2+)) conditions, whereas luminal pressure was varied between 25 and 150 mm Hg. The slope of the relationship between tone and pressure in the middle cerebral artery was 0.08±0.01 in control rats and was similar in normal pregnant rats (0.05±0.01). In the RUPP model of placental ischemia, this relationship was markedly reduced (slope=0.01±0.00; P<0.05). Endothelial dependent and independent dilation was not different between groups, nor was there evidence of vascular remodeling assessed by the wall:lumen ratio and calculated wall stress. The impaired myogenic response was associated with brain edema measured by percentage of water content (RUPP P<0.05 versus control and normal pregnant rats). This study demonstrates that placental ischemia in pregnant rats leads to impaired myogenic tone in the middle cerebral arteries and that the RUPP model is a potentially important tool to examine mechanisms leading to encephalopathy during preeclamptic pregnancies.
Collapse
Affiliation(s)
- Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 39047, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ogge G, Chaiworapongsa T, Romero R, Hussein Y, Kusanovic JP, Yeo L, Kim CJ, Hassan SS. Placental lesions associated with maternal underperfusion are more frequent in early-onset than in late-onset preeclampsia. J Perinat Med 2011; 39:641-52. [PMID: 21848483 PMCID: PMC3213694 DOI: 10.1515/jpm.2011.098] [Citation(s) in RCA: 198] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Preeclampsia (PE) has been classified into early- and late-onset disease. These two phenotypic variants of PE have been proposed to have a different pathophysiology. However, the gestational age cut-off to define "early" vs. "late" PE has varied among studies. The objective of this investigation was to determine the prevalence of lesions consistent with maternal underperfusion of the placenta in patients with PE as a function of gestational age. STUDY DESIGN A nested case-control study of 8307 singleton pregnant women who deliver after 20 weeks of gestation was constructed based on a cohort. Cases were defined as those with PE (n=910); controls were pregnant women who did not have a hypertensive disorder in pregnancy (n=7397). The frequency of maternal underperfusion of the placenta (according to the criteria of the Society for Pediatric Pathology) was compared between the two groups. Logistic regression was used for analysis. Estimated relative risks (RRs) were calculated from odds ratios. RESULTS 1) The prevalence of lesions consistent with maternal underperfusion was higher in patients with PE than in the control group [43.3% vs. 15.9%, unadjusted odds ratio 4.0 (95% CI 3.5-4.7); P<0.001]; 2) the estimated RR of maternal underperfusion lesions in PE was higher than in the control group [RR=2.8 (95% CI 2.5-3.0)]; 3) the lower the gestational age at delivery, the higher the RR for these lesions; 4) early-onset PE, regardless of the gestational age used to define it (<32, 33, 34, 35 or 37 weeks) had a significantly higher frequency of placental lesions consistent with maternal underperfusion than late-onset PE (P<0.001 for all). CONCLUSIONS 1) The earlier the gestational age of preeclampsia at delivery, the higher the frequency of placental lesions consistent with maternal underperfusion; 2) our data suggest that demonstrable placental involvement as determined by pathologic examination differs in early- and late-onset preeclampsia; and 3) this phenomenon appears to be a continuum, and we could not identify a clear and unambiguous gestational age at which lesions consistent with underperfusion would not be present.
Collapse
Affiliation(s)
- Giovanna Ogge
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA
| | - Youssef Hussein
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA
| | - Juan Pedro Kusanovic
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile and Center for Perinatal Research, Sótero del Río Hospital, Santiago, Chile
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Pathology, Wayne State University, Detroit, MI, USA
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
50
|
Hu XQ, Xiao D, Zhu R, Huang X, Yang S, Wilson S, Zhang L. Pregnancy upregulates large-conductance Ca(2+)-activated K(+) channel activity and attenuates myogenic tone in uterine arteries. Hypertension 2011; 58:1132-9. [PMID: 22042813 DOI: 10.1161/hypertensionaha.111.179952] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Uterine vascular tone significantly decreases whereas uterine blood flow dramatically increases during pregnancy. However, the complete molecular mechanisms remain elusive. We hypothesized that increased Ca(2+)-activated K(+) (BK(Ca)) channel activity contributes to the decreased myogenic tone of uterine arteries in pregnancy. Resistance-sized uterine arteries were isolated from nonpregnant and near-term pregnant sheep. Electrophysiological studies revealed a greater whole-cell K(+) current density in pregnant compared with nonpregnant uterine arteries. Tetraethylammonium and iberiotoxin inhibited K(+) currents to the same extent in uterine arterial myocytes. The BK(Ca) channel current density was significantly increased in pregnant uterine arteries. In accordance, tetraethylammonium significantly increased pressure-induced myogenic tone in pregnant uterine arteries and abolished the difference in myogenic responses between pregnant and nonpregnant uterine arteries. Activation of protein kinase C produced a similar effect to tetraethylammonium by inhibiting BK(Ca) channel activity and increasing myogenic tone in pregnant uterine arteries. Chronic treatment of nonpregnant uterine arteries with physiologically relevant concentrations of 17β-estradiol and progesterone caused a significant increase in the BK(Ca) channel current density. Western blot analyses demonstrated a significant increase of the β1, but not α, subunit of BK(Ca) channels in pregnant uterine arteries. In accordance, steroid treatment of nonpregnant uterine arteries resulted in an upregulation of the β1, but not α, subunit expression. The results indicate that the steroid hormone-mediated upregulation of the β1 subunit and BK(Ca) channel activity may play a key role in attenuating myogenic tone of the uterine artery in pregnancy.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | | | |
Collapse
|