1
|
Smith J, Powell M, Cromartie W, Smith S, Jones K, Castillo A, Shaw J, Editone J, Howard A, Tatum R, Smith A, Fisher B, Booz GW, Cunningham M. Intrauterine growth-restricted pregnant rats, from placental ischemic dams, display preeclamptic-like symptoms: A new rat model of preeclampsia. Physiol Rep 2024; 12:e70112. [PMID: 39482843 PMCID: PMC11527824 DOI: 10.14814/phy2.70112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
Preeclampsia (PE) is characterized by de novo hypertension (HTN) and is often associated with intrauterine growth restriction (IUGR). Hallmarks of PE are placental ischemia, decreased nitric oxide (NO) bioavailability, oxidative stress (OS), and organ damage in the kidneys and brain. This study aims to characterize a new model of PE using pregnant IUGR rats from hypertensive placental ischemic dams. It is hypothesized that pregnant IUGR rats from hypertensive placental ischemic dams will have elevated blood pressure (BP), OS, and organ damage. In this study, pregnant rats are divided into two groups: normal pregnant (NP) and hypertensive placental ischemic dams (RUPP). Offspring from NP and RUPP dams were mated at 10 weeks of age to generate pregnant IUGR (IUGR Preg) and pregnant control (CON Preg) rats. BP and other markers of PE were evaluated during late gestation. Pregnant IUGR rats had elevated BP and systemic OS. The maternal body weight of pregnant IUGR rats and their pups' weights were decreased, while the brains were enlarged with elevated OS. In summary, pregnant IUGR rats, born from hypertensive placental ischemic dams, have HTN and increased systemic and brain OS, with larger brain sizes and smaller pups. Furthermore, this study shows that pregnant IUGR rats exhibit a preeclamptic-like phenotype, suggesting a new epigenetic model of PE.
Collapse
Affiliation(s)
- Jonna Smith
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Madison Powell
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Whitney Cromartie
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Savanna Smith
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Kylie Jones
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Angie Castillo
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Jordan Shaw
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Joseph Editone
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Ahfiya Howard
- School of Social WorkTexas A & M University‐CommerceCommerceTexasUSA
| | - Robert Tatum
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Alex Smith
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Brandon Fisher
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - George W. Booz
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Mark Cunningham
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
2
|
Issotina Zibrila A, Zhou J, Wang X, Zeng M, Ali MA, Liu X, Alkuhali AA, Zeng Z, Meng Y, Wang Z, Li X, Liu J. Placental ischemia-upregulated angiotensin II type 1 receptor in hypothalamic paraventricular nucleus contributes to hypertension in rat. Pflugers Arch 2024; 476:1677-1691. [PMID: 39215834 DOI: 10.1007/s00424-024-03010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/30/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Preeclampsia (PE) is associated with increased angiotensin II sensitivity and poor neurological outcomes marked by temporal loss of neural control of blood pressure. Yet the role of centrally expressed angiotensin II type 1 receptor (AT1R) within the paraventricular nucleus of the hypothalamus (PVN) in the PE model is not understood. In a PE rat model with reduced placental perfusion pressure (RUPP) induced on gestational day 14 (GD14), the PVN expression and cellular localization of AT1R were assessed using immunofluorescence and western blotting. The sensitivity of RUPP to acute angiotensin II infusion was assessed. AT1R was antagonized by losartan (100 µg/kg/day) for 5 days intracerebroventricularly (ICV). Hemodynamic data and samples were collected on GD19 for further analysis. RUPP upregulated (p < 0.05) mRNA and protein of AT1R within the PVN and lowered (p < 0.05) circulating angiotensin II in rats. RUPP increased neural and microglial activation. Cellular localization assessment revealed that AT1R was primarily expressed in neurons and slightly in microglia and astrocytes. Infusion of 100 ng/kg as bolus increased the mean arterial pressure (MAP in mmHg) in both RUPP and Sham. ICV losartan infusion attenuated RUPP-increased MAP (113.6 ± 6.22 in RUPP vs. 92.16 ± 5.30 in RUPP + Los, p = 0.021) and the expression of nuclear transcription factor NF-κB, tyrosine hydroxylase (TH), NADPH oxidase 4 (NOX4) and reactive oxygen species (ROS) in the PVN. Our data suggest that centrally expressed AT1R, within the PVN, contributes to placental ischemia-induced hypertension in RUPP rats highlighting its therapeutic potential in PE.
Collapse
Affiliation(s)
- Abdoulaye Issotina Zibrila
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Jun Zhou
- Department of Pharmacology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an, 710061, Shaanxi, P. R. China
| | - Xiaomin Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Ming Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Md Ahasan Ali
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Xiaoxu Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Asma A Alkuhali
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Zhaoshu Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Yuan Meng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China
| | - Zheng Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Xuelan Li
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China.
| | - Jinjun Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, P. R. China.
| |
Collapse
|
3
|
Traynor M. Lung-protective ventilation in the management of congenital diaphragmatic hernia. WORLD JOURNAL OF PEDIATRIC SURGERY 2024; 7:e000789. [PMID: 39119150 PMCID: PMC11308893 DOI: 10.1136/wjps-2024-000789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Prioritizing lung-protective ventilation has produced a clear mortality benefit in neonates with congenital diaphragmatic hernia (CDH). While there is a paucity of CDH-specific evidence to support any particular approach to lung-protective ventilation, a growing body of data in adults is beginning to clarify the mechanisms behind ventilator-induced lung injury and inform safer management of mechanical ventilation in general. This review summarizes the adult data and attempts to relate the findings, conceptually, to the CDH population. Critical lessons from the adult studies are that much of the damage done during conventional mechanical ventilation affects normal lung tissue and that most of this damage occurs at the low-volume and high-volume extremes of the respiratory cycle. Consequently, it is important to prevent atelectasis by using sufficient positive end-expiratory pressure while also avoiding overdistention by scaling tidal volume to the amount of functional lung tissue rather than body weight. Paralysis early in acute respiratory distress syndrome improves outcomes, possibly because consistent respiratory mechanics facilitate avoidance of both atelectasis and overdistention-a mechanism that may also apply to the CDH population. Volume-targeted conventional modes may be advantageous in CDH, but determining optimal tidal volume is challenging. Both high-frequency oscillatory ventilation and high-frequency jet ventilation have been used successfully as 'rescue modes' to avoid extracorporeal membrane oxygenation, and a prospective trial comparing the two high-frequency modalities as the primary ventilation strategy for CDH is underway.
Collapse
Affiliation(s)
- Mike Traynor
- Department of Anesthesia, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Fitzgerald S, Deer E, Hogg J, Cornelius DC, Turner T, Amaral LM, Hoang N, Edwards K, Herrock O, Campbell N, Ibrahim T, LaMarca B. RUPP Th17s cause hypertension and mitochondrial dysfunction in the kidney and placenta during pregnancy. Pregnancy Hypertens 2023; 32:50-56. [PMID: 37104924 PMCID: PMC11494691 DOI: 10.1016/j.preghy.2023.04.002] [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: 12/27/2022] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND Preeclampsia (PE), new-onset hypertension (HTN), and organ dysfunction during the second half of pregnancy, is associated with an increase in inflammatory immune cells, including T helper 17 (Th17) cells. Studies have demonstrated that mitochondrial (mt) dysfunction is important in the pathogenesis of PE though causative factors have yet to be fully identified. Although Th17 cells, natural killer (NK) cells, and mt dysfunction contribute to HTN in the reduced uterine perfusion pressure (RUPP) rat model, the role of Th17 cells or IL-17 in mt dysfunction is unknown. Therefore, we hypothesize that RUPP stimulated Th17 cells cause HTN and mt dysfunction, which is alleviated with the blockade of IL-17. METHODS On gestational day 12 (GD12), RUPP Th17 cells were transferred into normal pregnant (NP) Sprague Dawley rats. A subset of NP + RUPPTh17 rats received IL-17RC (100 pg/day) on GD14-19. Blood pressure (MAP), NK cells, and mt function were measured on GD19 in all groups. RESULTS MAP increased in response to NP + RUPP Th17 compared to NP rats and was lowered with IL-17RC. Circulating and placental NK cells increased with NP + RUPP Th17 compared to NP and were lowered with IL-17RC. Renal mtROS increased in NP + RUPP Th17 compared to NP and was normalized with IL-17RC. Similar to PE women, placental mtROS decreased in NP + RUPP Th17 and was normalized with IL-17RC. CONCLUSION Our results indicate that IL-17RC inhibition normalizes HTN, NK cell activation, and multi-organ mt dysfunction caused by Th17 cells stimulated in response to placental ischemia.
Collapse
Affiliation(s)
- Sarah Fitzgerald
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - James Hogg
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Denise C Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ngoc Hoang
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kristin Edwards
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, United States; Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States.
| |
Collapse
|
5
|
Escudero C, Kupka E, Ibañez B, Sandoval H, Troncoso F, Wikström AK, López-Espíndola D, Acurio J, Torres-Vergara P, Bergman L. Brain Vascular Dysfunction in Mothers and Their Children Exposed to Preeclampsia. Hypertension 2023; 80:242-256. [PMID: 35920147 DOI: 10.1161/hypertensionaha.122.19408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Preeclampsia is a maternal syndrome characterized by the new onset of hypertension and proteinuria after 20 weeks of gestation associated with multisystemic complications, including brain alterations. Indeed, brain complications associated with preeclampsia are the leading direct causes of fetal and maternal morbidity and mortality, especially in low- and middle-income countries. In addition to the well-recognized long-term adverse cardiovascular effects of preeclampsia, women who have had preeclampsia have higher risk of stroke, dementia, intracerebral white matter lesions, epilepsy, and perhaps also cognitive decline postpartum. Furthermore, increasing evidence has also associated preeclampsia with similar cognitive and cerebral disorders in the offspring. However, the mechanistic links between these associations remain unresolved. This article summarizes the current knowledge about the cerebrovascular complications elicited by preeclampsia and the potential pathophysiological mechanisms involved, emphasizing the impaired brain vascular function in the mother and their offspring.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Ellen Kupka
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Sweden (E.K.)
| | - Belen Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.)
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, University of Bío-Bío, Chillán, Chile (C.E., B.I., H.S., F.T., J.A.).,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile (C.E., J.A., P.T.-V.).,Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Chile (P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (A.K.W., L.B.).,Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.).,Department of clinical sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
| |
Collapse
|
6
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Carrasco-Wong I, González-Ortiz M, Araujo GG, Lima VV, Giachini FR, Stojanova J, Moller A, Martín SS, Escudero P, Damiano AE, Sosa-Macias M, Galaviz-Hernandez C, Teran E, Escudero C. The Placental Function Beyond Pregnancy: Insights from Latin America. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:287-307. [PMID: 37466779 DOI: 10.1007/978-3-031-32554-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Currently, more than 100,000 papers had been published studying the placenta in both physiological and pathological contexts. However, relevant health conditions affecting placental function, mostly found in low-income countries, should be evaluated deeper. This review will raise some - of what we think necessary - points of discussion regarding challenging topics not fully understood, including the paternal versus maternal contribution on placental genes imprinting, placenta-brain communication, and some environmental conditions affecting the placenta. The discussions are parts of an international effort to fulfil some gaps observed in this area, and Latin-American research groups currently evaluate that.
Collapse
Affiliation(s)
- Ivo Carrasco-Wong
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
| | - Gabriel Gomes Araujo
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidade Federal de Mato Grosso, Barra do Garcas, Brazil
| | - Victor V Lima
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidade Federal de Mato Grosso, Barra do Garcas, Brazil
| | - Fernanda R Giachini
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidade Federal de Mato Grosso, Barra do Garcas, Brazil
| | - Jana Stojanova
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
| | - Alejandra Moller
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Sebastián San Martín
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
| | - Pablo Escudero
- Faculty of Medicine, Universidad San Sebastian, Sede Concepcion, Chile
| | - Alicia E Damiano
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)- CONICET- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martha Sosa-Macias
- Genomics Academia, Instituto Politécnico Nacional-CIIDIR Durango, Durango, Mexico
| | | | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Quito, Ecuador
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
- Vascular Physiology Laboratory, Basic Sciences Department, Faculty of Sciences, Universidad del Bio-Bio, Chillan, Chile.
| |
Collapse
|
8
|
Misan N, Michalak S, Rzymski P, Poniedziałek B, Kapska K, Osztynowicz K, Ropacka-Lesiak M. Molecular Indicators of Blood-Brain Barrier Breakdown and Neuronal Injury in Pregnancy Complicated by Fetal Growth Restriction. Int J Mol Sci 2022; 23:ijms232213798. [PMID: 36430274 PMCID: PMC9695431 DOI: 10.3390/ijms232213798] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
This study evaluated the damage to the endothelial tight junctions (TJs) in pregnancies complicated by fetal growth restriction (FGR) and investigated whether FGR is related to blood-brain barrier disintegration and, subsequently, to the appearance of proteins indicative of neuronal injury in maternal blood. The studied group included 90 pregnant women diagnosed with FGR. The control group consisted of 70 women with an uncomplicated pregnancy. The biochemical measurements included serum neuronal proteins (subunit of the N-methyl-D-aspartate receptor-NR1, nucleoside diphosphate kinase A-NME1, and S100 calcium-binding protein B-S100B), serum TJ proteins (occludin-OCLN, claudin-5-CLN5, zonula occludens-zo-1, and OCLN/zo-1 and CLN5/zo-1 ratios), and placental expression of TJ proteins (OCLN, claudin-4 CLN4, CLN5, zo-1). The significantly higher serum S100B and CLN5 levels and serum CLN5/zo-1 ratio were observed in FGR compared to healthy pregnancies. Moreover, FGR was characterized by increased placental CLN5 expression. Both serum NME1 levels and placental CLN4 expression in FGR pregnancies were significantly related to the incidence of neurological disorders in newborns. Mothers of FGR neonates who developed neurological complications and intraventricular hemorrhage (IVH) had statistically higher NME1 concentrations during pregnancy and significantly lower placental CLN4 expression than mothers of FGR neonates without neurological abnormalities. The serum NME1 levels and placental CLN4 expression were predictive markers of IVH in the FGR group. The blood-brain barrier is destabilized in pregnancies complicated by FGR. Neurological disorders, including IVH, are associated with higher serum concentrations of NME1 and the decreased placental expression of CLN4. The serum NME1 levels and placental CLN4 expression may serve as biomarkers, helpful in predicting IVH in FGR. It may allow for more precise monitoring and influence decision-making on the optimal delivery time to avoid developing neurological complications.
Collapse
Affiliation(s)
- Natalia Misan
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
- Correspondence:
| | - Sławomir Michalak
- Department of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-848 Poznan, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| | - Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Katarzyna Kapska
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Krystyna Osztynowicz
- Department of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mariola Ropacka-Lesiak
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| |
Collapse
|
9
|
Canjels LPW, Jansen JFA, Alers RJ, Ghossein‐Doha C, van den Kerkhof M, Schiffer VMMM, Mulder E, Gerretsen SC, Aldenkamp AP, Hurks PPM, van de Ven V, Spaanderman MEA, Backes WH. Blood-brain barrier leakage years after pre-eclampsia: dynamic contrast-enhanced 7-Tesla MRI study. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 60:541-548. [PMID: 35502137 PMCID: PMC9826493 DOI: 10.1002/uog.24930] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Pre-eclampsia is a hypertensive complication of pregnancy that is associated with an increased risk of long-term cardiovascular and cerebrovascular disorders. Although the underlying mechanism of persistent susceptibility to cerebral complications after pre-eclampsia remains largely unclear, impaired blood-brain barrier (BBB) integrity has been suggested to precede several cerebrovascular diseases. In this study, we aimed to investigate the integrity of the BBB years after pre-eclampsia. METHODS This was an observational study of premenopausal formerly pre-eclamptic women and controls with a history of normotensive pregnancy who underwent cerebral magnetic resonance imaging (MRI) at ultra-high field (7 Tesla) to assess the integrity of the BBB. Permeability of the BBB was determined by assessing leakage rate and fractional leakage volume of the contrast agent gadobutrol using dynamic contrast-enhanced MRI. BBB leakage measures were determined for the whole brain and lobar white and gray matter. Multivariable analyses were performed, and odds ratios were calculated to compare women with and those without a history of pre-eclampsia, adjusting for potential confounding effects of age, hypertension status at MRI and Fazekas score. RESULTS Twenty-two formerly pre-eclamptic women (mean age, 37.8 ± 5.4 years) and 13 control women with a history of normotensive pregnancy (mean age, 40.8 ± 5.5 years) were included in the study. The time since the index pregnancy was 6.6 ± 3.2 years in the pre-eclamptic group and 9.0 ± 3.7 years in controls. The leakage rate and fractional leakage volume were significantly higher in formerly pre-eclamptic women than in controls in the global white (P = 0.001) and gray (P = 0.02) matter. Regionally, the frontal (P = 0.04) and parietal (P = 0.009) cortical gray matter, and the frontal (P = 0.001), temporal (P < 0.05) and occipital (P = 0.007) white matter showed higher leakage rates in formerly pre-eclamptic women. The odds of a high leakage rate after pre-eclampsia were generally higher in white-matter regions than in gray-matter regions. CONCLUSION This observational study demonstrates global impairment of the BBB years after a pre-eclamptic pregnancy, which could be an early marker of long-term cerebrovascular disorders. © 2022 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- L. P. W. Canjels
- Department of Radiology & Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
- MHeNs, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - J. F. A. Jansen
- Department of Radiology & Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
- MHeNs, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Department of Electrical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | - R. J. Alers
- Department of Gynaecology and ObstetricsMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
- GROW, School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - C. Ghossein‐Doha
- GROW, School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
- CARIM, School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
- Department of CardiologyMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
| | - M. van den Kerkhof
- Department of Radiology & Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
- MHeNs, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - V. M. M. M. Schiffer
- Department of Gynaecology and ObstetricsMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
- GROW, School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - E. Mulder
- Department of Gynaecology and ObstetricsMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
- GROW, School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - S. C. Gerretsen
- Department of Radiology & Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - A. P. Aldenkamp
- MHeNs, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Department of Electrical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
- Academic Center for Epileptology Kempenhaeghe/ Maastricht University Medical Center (MUMC+)Heeze and MaastrichtThe Netherlands
- Department of NeurologyMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
| | - P. P. M. Hurks
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - V. van de Ven
- Department of Cognitive Neuroscience, Faculty of Psychology and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - M. E. A. Spaanderman
- Department of Gynaecology and ObstetricsMaastricht University Medical Center (MUMC+)MaastrichtThe Netherlands
- GROW, School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - W. H. Backes
- Department of Radiology & Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
- MHeNs, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- CARIM, School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
10
|
Nemeth Z, Granger JP, Ryan MJ, Drummond HA. Is there a role of proinflammatory cytokines on degenerin-mediated cerebrovascular function in preeclampsia? Physiol Rep 2022; 10:e15376. [PMID: 35831968 PMCID: PMC9279847 DOI: 10.14814/phy2.15376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/25/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023] Open
Abstract
Preeclampsia (PE) is associated with adverse cerebrovascular effects during and following parturition including stroke, small vessel disease, and vascular dementia. A potential contributing factor to the cerebrovascular dysfunction is the loss of cerebral blood flow (CBF) autoregulation. Autoregulation is the maintenance of CBF to meet local demands with changes in perfusion pressure. When perfusion pressure rises, vasoconstriction of cerebral arteries and arterioles maintains flow and prevents the transfer of higher systemic pressure to downstream microvasculature. In the face of concurrent hypertension, loss of autoregulatory control exposes small delicate microvessels to injury from elevated systemic blood pressure. While placental ischemia is considered the initiating event in the preeclamptic cascade, the factor(s) mediating cerebrovascular dysfunction are poorly understood. Elevated plasma proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin-17 (IL-17), are potential mediators of autoregulatory loss. Impaired CBF responses to increases in systemic pressure are attributed to the impaired pressure-induced (myogenic) constriction of small cerebral arteries and arterioles in PE. Myogenic vasoconstriction is initiated by pressure-induced vascular smooth muscle cell (VSMC) stretch. Recent studies from our laboratory group indicate that proinflammatory cytokines impair the myogenic mechanism of CBF autoregulation via inhibition of vascular degenerin proteins, putative mediators of myogenic constriction in VSMCs. This brief review links studies showing the effect of proinflammatory cytokines on degenerin expression and CBF autoregulation to the pathological cerebral consequences of preeclampsia.
Collapse
Affiliation(s)
- Zoltan Nemeth
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Institute of Translational MedicineFaculty of Medicine, Semmelweis UniversityBudapestHungary
- Department of Morphology and PhysiologyFaculty of Health Sciences, Semmelweis UniversityBudapestHungary
| | - Joey P. Granger
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Michael J. Ryan
- Department of Pharmacology, Physiology and NeuroscienceUniversity of South Carolina School of MedicineColumbiaSouth CarolinaUSA
| | - Heather A. Drummond
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
11
|
Torres-Vergara P, Rivera R, Escudero C. How Soluble Fms-Like Tyrosine Kinase 1 Could Contribute to Blood-Brain Barrier Dysfunction in Preeclampsia? Front Physiol 2022; 12:805082. [PMID: 35211027 PMCID: PMC8862682 DOI: 10.3389/fphys.2021.805082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Preeclampsia is a pregnancy-related syndrome that courses with severe cerebrovascular complications if not properly managed. Findings from pre-clinical and clinical studies have proposed that the imbalance between pro- and anti-angiogenic factors exhibited in preeclampsia is a major component of its pathophysiology. In this regard, measurement of circulating levels of soluble tyrosine kinase-1 similar to fms (sFlt-1), a decoy receptor for vascular endothelial growth factor (VEGF), is a moderately reliable biomarker for the diagnosis of preeclampsia. However, few studies have established a mechanistic approach to determine how the high levels of sFlt-1 are responsible for the endothelial dysfunction, and even less is known about its effects at the blood-brain barrier (BBB). Since the expression pattern of VEGF receptors type 1 and 2 in brain endothelial cells differs from the observed in peripheral endothelial cells, and components of the neurovascular unit of the BBB provide paracrine secretion of VEGF, this compartmentalization of VEGF signaling could help to see in a different viewpoint the role of sFlt-1 in the development of endothelial dysfunction. In this article, we provide a hypothesis of how sFlt-1 could eventually be a protective factor for brain endothelial cells of the BBB under preeclampsia.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Department of Pharmacy, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Robin Rivera
- Department of Pharmacy, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile
| |
Collapse
|
12
|
Friis T, Wikström AK, Acurio J, León J, Zetterberg H, Blennow K, Nelander M, Åkerud H, Kaihola H, Cluver C, Troncoso F, Torres-Vergara P, Escudero C, Bergman L. Cerebral Biomarkers and Blood-Brain Barrier Integrity in Preeclampsia. Cells 2022; 11:cells11050789. [PMID: 35269411 PMCID: PMC8909006 DOI: 10.3390/cells11050789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/10/2022] [Accepted: 02/20/2022] [Indexed: 01/25/2023] Open
Abstract
Cerebral complications in preeclampsia contribute substantially to maternal mortality and morbidity. There is a lack of reliable and accessible predictors for preeclampsia-related cerebral complications. In this study, plasma from women with preeclampsia (n = 28), women with normal pregnancies (n = 28) and non-pregnant women (n = 16) was analyzed for concentrations of the cerebral biomarkers neurofilament light (NfL), tau, neuron-specific enolase (NSE) and S100B. Then, an in vitro blood−brain barrier (BBB) model, based on the human cerebral microvascular endothelial cell line (hCMEC/D3), was employed to assess the effect of plasma from the three study groups. Transendothelial electrical resistance (TEER) was used as an estimation of BBB integrity. NfL and tau are proteins expressed in axons, NSE in neurons and S100B in glial cells and are used as biomarkers for neurological injury in other diseases such as dementia, traumatic brain injury and hypoxic brain injury. Plasma concentrations of NfL, tau, NSE and S100B were all higher in women with preeclampsia compared with women with normal pregnancies (8.85 vs. 5.25 ng/L, p < 0.001; 2.90 vs. 2.40 ng/L, p < 0.05; 3.50 vs. 2.37 µg/L, p < 0.001 and 0.08 vs. 0.05 µg/L, p < 0.01, respectively). Plasma concentrations of NfL were also higher in women with preeclampsia compared with non-pregnant women (p < 0.001). Higher plasma concentrations of the cerebral biomarker NfL were associated with decreased TEER (p = 0.002) in an in vitro model of the BBB, a finding which indicates that NfL could be a promising biomarker for BBB alterations in preeclampsia.
Collapse
Affiliation(s)
- Therese Friis
- Department of Women’s and Children’s Health, Uppsala University, 75185 Uppsala, Sweden; (A.-K.W.); (M.N.); (L.B.)
- Correspondence: ; Tel.: +46-18-611-6613
| | - Anna-Karin Wikström
- Department of Women’s and Children’s Health, Uppsala University, 75185 Uppsala, Sweden; (A.-K.W.); (M.N.); (L.B.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán 3810178, Chile; (J.A.); (J.L.); (F.T.); (C.E.)
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3810178, Chile;
| | - José León
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán 3810178, Chile; (J.A.); (J.L.); (F.T.); (C.E.)
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3810178, Chile;
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Los Ángeles 4441171, Chile
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 43180 Mölndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 43180 Mölndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
| | - Maria Nelander
- Department of Women’s and Children’s Health, Uppsala University, 75185 Uppsala, Sweden; (A.-K.W.); (M.N.); (L.B.)
| | - Helena Åkerud
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (H.Å.); (H.K.)
| | - Helena Kaihola
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (H.Å.); (H.K.)
| | - Catherine Cluver
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town 7500, South Africa;
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán 3810178, Chile; (J.A.); (J.L.); (F.T.); (C.E.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3810178, Chile;
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán 3810178, Chile; (J.A.); (J.L.); (F.T.); (C.E.)
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3810178, Chile;
| | - Lina Bergman
- Department of Women’s and Children’s Health, Uppsala University, 75185 Uppsala, Sweden; (A.-K.W.); (M.N.); (L.B.)
- Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town 7500, South Africa;
- Department of Obstetrics and Gynecology, Gothenburg University, 41650 Gothenburg, Sweden
| |
Collapse
|
13
|
Abstract
Women face a disproportionate burden of stroke mortality and disability. Biologic sex and sociocultural gender both contribute to differences in stroke risk factors, assessment, treatment, and outcomes. There are substantial differences in the strength of association of stroke risk factors, as well as female-specific risk factors. Moreover, there are differences in presentation, response to treatment, and stroke outcomes in women. This review outlines current knowledge of impact of sex and gender on stroke, as well as delineates research gaps and areas for future inquiry.
Collapse
Affiliation(s)
- Kathryn M. Rexrode
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Tracy E. Madsen
- Division of Sex and Gender in Emergency Medicine, Department of Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, RI; Department of Epidemiology, Brown University School of Public Health, Providence RI
| | - Amy Y. X. Yu
- Department of Medicine (Neurology), University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Cheryl Carcel
- Neurology Program, The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Judith H. Lichtman
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Eliza C. Miller
- Division of Stroke and Cerebrovascular Disease, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
14
|
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
|
15
|
Evidence of Neuroinflammation and Blood-Brain Barrier Disruption in Women with Preeclampsia and Eclampsia. Cells 2021; 10:cells10113045. [PMID: 34831266 PMCID: PMC8616341 DOI: 10.3390/cells10113045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/16/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Cerebral complications in preeclampsia are leading causes of maternal mortality. Animal models suggest that an injured blood-brain barrier and neuroinflammation may be important but there is paucity of data from human studies. Therefore, we aimed to evaluate this in women with preeclampsia and eclampsia. We included women recruited to the South African Preeclampsia Obstetric Adverse Events (PROVE) biobank. Blood and cerebrospinal fluid (CSF) were collected around delivery. CSF was analyzed for neuroinflammatory markers interleukin 1β, interleukin 6, interleukin-8 and tumor necrosis factor alpha (TNF-alpha). The CSF to plasma albumin ratio was measured to assess blood-brain barrier function. Women with eclampsia (n = 4) showed increased CSF concentrations of all pro-inflammatory cytokines and TNF-alpha compared to women with normotensive pregnancies (n = 7) and also for interleukin-6 and TNF-alpha compared to women with preeclampsia (n = 4). Women with preeclampsia also showed increases in pro-inflammatory cytokines IL-6 and IL-8 but not TNF-alpha in the CSF compared to women with normotensive pregnancies. In particular, women with eclampsia but also women with preeclampsia showed an increase in the CSF to plasma albumin ratio compared to normotensive women. In conclusion, women with preeclampsia and eclampsia show evidence of neuroinflammation and an injured blood-brain barrier. These findings are seen in particular among women with eclampsia.
Collapse
|
16
|
León J, Acurio J, Bergman L, López J, Karin Wikström A, Torres-Vergara P, Troncoso F, Castro FO, Vatish M, Escudero C. Disruption of the Blood-Brain Barrier by Extracellular Vesicles From Preeclampsia Plasma and Hypoxic Placentae: Attenuation by Magnesium Sulfate. Hypertension 2021; 78:1423-1433. [PMID: 34601964 DOI: 10.1161/hypertensionaha.121.17744] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- José León
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile (J. Leon,.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
- Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
- Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.)
| | - Juán López
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
| | - Anna Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Chile (P.T.-V.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepcion, Chillan, Chile (F.O.C.)
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom (M.V.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| |
Collapse
|
17
|
Liew J, Feghali J, Huang J. Intracerebral and subarachnoid hemorrhage in pregnancy. HANDBOOK OF CLINICAL NEUROLOGY 2021; 172:33-50. [PMID: 32768093 DOI: 10.1016/b978-0-444-64240-0.00002-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Maternal stroke occurs in around 34 out of every 100,000 deliveries and is responsible for around 5%-12% of all maternal deaths. It is most commonly hemorrhagic, and women are at highest risk for developing pregnancy-related hemorrhage during the early postpartum period through 6 weeks following the delivery. The most common causes of hemorrhagic stroke in pregnant patients are arteriovenous malformations and cerebral aneurysms. Management is similar to that for acute hemorrhagic stroke in the nonpregnant population with standard use of computed tomography and judicious utilization of intracranial vessel imaging and contrast. The optimal delivery method is evaluated on a case-by-case basis, and cesarean delivery is not always required. As most current studies are limited by retrospective design, relatively small sample sizes, and heterogeneous study term definitions, strong and comprehensive evidence-based guidelines on the management of acute hemorrhagic stroke in pregnant patients are still lacking. In the future, multicenter registries and prospective studies with uniform definitions will help improve management strategies in this complex patient population.
Collapse
Affiliation(s)
- Jason Liew
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - James Feghali
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Judy Huang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
18
|
Duncan JW, Nemeth Z, Hildebrandt E, Granger JP, Ryan MJ, Drummond HA. Interleukin-17 induces hypertension but does not impair cerebrovascular function in pregnant rats. Pregnancy Hypertens 2021; 24:50-57. [PMID: 33677419 PMCID: PMC8159853 DOI: 10.1016/j.preghy.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/20/2021] [Accepted: 02/16/2021] [Indexed: 11/24/2022]
Abstract
Preeclampsia affects 5-8% of pregnancies and is characterized by hypertension, placental ischemia, neurological impairment, and an increase in circulating inflammatory cytokines, including Interleukin-17 (IL17). While placental ischemia has also been shown to impair cerebrovascular function, it is not known which placental-associated factor(s) drive this effect. The purpose of this study was to examine the effects of IL17 on cerebrovascular function during pregnancy. To achieve this goal, pregnant rats were infused with either IL17 (150 pg/day, 5 days, osmotic minipump), or vehicle (saline/0.7% BSA osmotic minipump) starting at gestational day (GD) 14. On GD 19, the cerebral blood flow (CBF) response to increases in mean arterial pressure (MAP) was measured in vivo, and myogenic constrictor responses of the middle cerebral artery (MCA) were assessed ex vivo. IL17 increased MAP but impaired CBF responses only at the highest arterial pressure measured (190 mmHg). Myogenic constrictor responses overall were mostly unaffected by IL17 infusion; however, the intraluminal pressure at which peak myogenic tone was generated was lower in the IL17 infused group (120 vs 165 mm Hg), suggesting maximal tone is exerted at lower intraluminal pressures in IL17-treated pregnant rats. Consistent with the lack of substantial change in overall myogenic responsiveness, there was no difference in cerebral vessel expression of putative mechanosensitive protein βENaC, but a tendency towards a decrease in ASIC2 (p = 0.067) in IL17 rats. This study suggests that infusion of IL17 independent of other placental ischemia-associated factors is insufficient to recapitulate the features of impaired cerebrovascular function during placental ischemia. Further studies to examine of the role of other pro-inflammatory cytokines, individually or a combination, are necessary to determine mechanisms of cerebral vascular dysfunction during preeclampsia.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA
| | - Zoltan Nemeth
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA
| | - Emily Hildebrandt
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA
| | - Heather A Drummond
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39206, USA.
| |
Collapse
|
19
|
Maternal microvascular dysfunction during preeclamptic pregnancy. Clin Sci (Lond) 2021; 135:1083-1101. [PMID: 33960392 DOI: 10.1042/cs20200894] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy effecting ∼5-8% of pregnancies in the United States, and ∼8 million pregnancies worldwide. Preeclampsia is clinically diagnosed after the 20th week of gestation and is characterized by new onset hypertension accompanied by proteinuria and/or thrombocytopenia, renal insufficiency, impaired liver function, pulmonary edema, or cerebral or visual symptoms. This broad definition emphasizes the heterogeneity of the clinical presentation of preeclampsia, but also underscores the role of the microvascular beds, specifically the renal, cerebral, and hepatic circulations, in the pathophysiology of the disease. While the diagnostic criteria for preeclampsia relies on the development of de novo hypertension and accompanying clinical symptoms after 20-week gestation, it is likely that subclinical dysfunction of the maternal microvascular beds occurs in parallel and may even precede the development of overt cardiovascular symptoms in these women. However, little is known about the physiology of the non-reproductive maternal microvascular beds during preeclampsia, and the mechanism(s) mediating microvascular dysfunction during preeclamptic pregnancy are largely unexplored in humans despite their integral role in the pathophysiology of the disease. Therefore, the purpose of this review is to provide a summary of the existing literature on maternal microvascular dysfunction during preeclamptic pregnancy by reviewing the functional evidence in humans, highlighting potential mechanisms, and providing recommendations for future work in this area.
Collapse
|
20
|
Neurology of Preeclampsia and Related Disorders: an Update in Neuro-obstetrics. Curr Pain Headache Rep 2021; 25:40. [PMID: 33825997 PMCID: PMC10069269 DOI: 10.1007/s11916-021-00958-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Preeclampsia and related hypertensive disorders of pregnancy affect up to 10% of pregnancies. Neurological complications are common and neurologists often become involved in the care of obstetric patients with preeclampsia. Here, we review the definition(s), epidemiology, clinical features, and pathophysiology of preeclampsia, focusing on maternal neurological complications and headache as a common presenting symptom of preeclampsia. RECENT FINDINGS Neurological symptoms are early and disease-defining features of preeclampsia. Neurological complications of preeclampsia may include headaches, visual symptoms, cerebral edema, seizures, or acute cerebrovascular disorders such as intracerebral hemorrhage or reversible cerebral vasoconstriction syndrome. A history of migraine is an independent risk factor for vascular diseases during pregnancy, including preeclampsia and maternal stroke. The pathophysiology of both preeclampsia and migraine is complex, and the mechanisms linking the two are not fully understood. Overlapping clinical and pathophysiological features of migraine and preeclampsia include inflammation, vascular endothelial dysfunction, and changes in vasoreactivity. Neurological complications are recognized as a major contributor to maternal morbidity and mortality. Pregnant and postpartum women commonly present with headache, and red flags in the clinical history and examination should prompt urgent neuroimaging and laboratory evaluation. A focused headache history should be elicited from patients as part of routine obstetrical care to identify patients at an increased risk of preeclampsia and related hypertensive disorders of pregnancy. Collaborative models of care and scientific investigation in the emerging field of neuro-obstetrics have the common goal of reducing the risk of maternal neurological morbidity and mortality from preeclampsia.
Collapse
|
21
|
Bergman L, Acurio J, Leon J, Gatu E, Friis T, Nelander M, Wikström J, Larsson A, Lara E, Aguayo C, Torres-Vergara P, Wikström AK, Escudero C. Preeclampsia and Increased Permeability Over the Blood-Brain Barrier: A Role of Vascular Endothelial Growth Receptor 2. Am J Hypertens 2021; 34:73-81. [PMID: 32866228 DOI: 10.1093/ajh/hpaa142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/25/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cerebral complications in preeclampsia are leading causes of maternal mortality worldwide but pathophysiology is largely unknown and a challenge to study. Using an in vitro model of the human blood-brain barrier (BBB), we explored the role of vascular endothelial growth factor receptor 2 (VEGFR2) in preeclampsia. METHODS The human brain endothelial cell line (hCMEC/D3) cultured on Tranwells insert was exposed (12 hours) to plasma from women with preeclampsia (n = 28), normal pregnancy (n = 28), and nonpregnant (n = 16) controls. Transendothelial electrical resistance (TEER) and permeability to 70 kDa fluorescein isothiocyanate (FITC)-dextran were measured for the assessment of BBB integrity. We explored possible underlying mechanisms, with a focus on the expression of tight junction proteins and phosphorylation of 2 tyrosine residues of VEGFR2, associated with vascular permeability and migration (pY951) and cell proliferation (pY1175). Plasma concentrations of soluble FMS-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) were also measured. RESULTS hCMEC/D3 exposed to plasma from women with preeclampsia exhibited reduced TEER and increased permeability to 70 kDa FITC-dextran. These cells upregulated the messenger ribonucleic acid (mRNA) levels of VEGFR2, and pY951-VEGFR2, but reduced pY1175-VEGFR2 (P < 0.05 in all cases). No difference in mRNA expression of tight junction protein was observed between groups. There was no correlation between angiogenic biomarkers and BBB permeability. CONCLUSIONS We present a promising in vitro model of the BBB in preeclampsia. Selective tyrosine phosphorylation of VEGFR2 may participate in the increased BBB permeability in preeclampsia irrespective of plasma concentrations of angiogenic biomarkers.
Collapse
Affiliation(s)
- Lina Bergman
- Department of Women’s and Children’s Health, Uppsala University, Uppsala,Sweden
- Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Stellenbosch University, Stellenbosch, South Africa
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jose Leon
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile
| | - Emily Gatu
- Department of Women’s and Children’s Health, Uppsala University, Uppsala,Sweden
| | - Therese Friis
- Department of Women’s and Children’s Health, Uppsala University, Uppsala,Sweden
| | - Maria Nelander
- Department of Women’s and Children’s Health, Uppsala University, Uppsala,Sweden
| | - Johan Wikström
- Department of Radiology, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Claudio Aguayo
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Anna-Karin Wikström
- Department of Women’s and Children’s Health, Uppsala University, Uppsala,Sweden
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Sciences, University of Bío-Bío, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| |
Collapse
|
22
|
Wang S, Roman RJ, Fan F. Duration and magnitude of bidirectional fluctuation in blood pressure: the link between cerebrovascular dysfunction and cognitive impairment following spinal cord injury. JOURNAL OF NEUROBIOLOGY AND PHYSIOLOGY 2020; 2:15-18. [PMID: 33336208 PMCID: PMC7739907 DOI: 10.46439/neurobiology.2.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Individuals with spinal cord injury (SCI) have a significantly increased risk for cognitive impairment that is associated with cerebrovascular remodeling and endothelial dysfunction. The sub-acute stage following high thoracic SCI is characterized by increased fibrosis and stiffness of cerebral arteries. However, a more prolonged duration after SCI exacerbates cerebrovascular injury by damaging endothelium. Endothelial dysfunction is associated with reduced expression of transient receptor potential cation channel 4 that mediates the production of nitric oxide and epoxyeicosatrienoic acids following shear stress and the response to carbachol and other endothelium-dependent vasodilators. Reduced expression of CD31 in cerebral arteries also suggests the loss of endothelial cell integrity following chronic SCI. Repetitively transient hypertension and intermittent hypotension contribute to cerebrovascular endothelial dysfunction in the animals with a sub-acute stage of high thoracic SCI. The increase in vascular remodeling and endothelial dysfunction ultimately reduce cerebral blood flow, which promotes cerebral hypoperfusion and cognitive dysfunction in the chronic phase of SCI. In conclusion, the duration and magnitude of fluctuations in blood pressure after SCI play a vital role in the onset and progress of cerebrovascular dysfunction, which promotes the development of cognitive impairment.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
23
|
Neuropathophysiology of preeclampsia and eclampsia: A review of cerebral hemodynamic principles in hypertensive disorders of pregnancy. Pregnancy Hypertens 2020; 23:104-111. [PMID: 33310389 DOI: 10.1016/j.preghy.2020.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Preeclampsia and eclampsia are hypertensive disorders of pregnancy associated with abnormal placental vascular development. The systemic angiogenic imbalance, endothelial dysfunction and proinflammatory state caused by abnormal placental development results in abnormalities in renal, hepatic, pulmonary and neurologic function. Neurosensory symptoms related to pregnancy induced hypertension (PIH), the most devastating of which are intracranial hemorrhage and seizure, are among the leading causes of maternal and perinatal morbidity and mortality globally, yet risk stratification strategies and targeted therapies remain elusive. Current treatment for preeclampsia with severe features is limited to delivery, antihypertensive therapy, and magnesium sulfate seizure prophylaxis. Magnesium sulfate reduces seizure rates among severe preeclamptics, but predisposes patients to weakness, uterine atony, pulmonary edema and respiratory depression. Therefore, this drug should ideally be administered only to the subset of preeclamptics who are at increased risk for neurologic complications. While there are no objective methods validated to predict eclampsia, we hypothesize that measurement of optic nerve sheath diameters, optic disc height and middle cerebral artery transcranial doppler resistance indices may be useful in identifying subclinical cerebral edema, potentially allowing us to recognize those patients at highest risk for seizures. This summary of the current literature provides an initial framework for developing more sophisticated and noninvasive methods for identifying, monitoring and treating parturients who are at highest risk for neurologic complications from preeclampsia.
Collapse
|
24
|
Bakrania BA, Spradley FT, Drummond HA, LaMarca B, Ryan MJ, Granger JP. Preeclampsia: Linking Placental Ischemia with Maternal Endothelial and Vascular Dysfunction. Compr Physiol 2020; 11:1315-1349. [PMID: 33295016 PMCID: PMC7959189 DOI: 10.1002/cphy.c200008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Preeclampsia (PE), a hypertensive disorder, occurs in 3% to 8% of pregnancies in the United States and affects over 200,000 women and newborns per year. The United States has seen a 25% increase in the incidence of PE, largely owing to increases in risk factors, including obesity and cardiovascular disease. Although the etiology of PE is not clear, it is believed that impaired spiral artery remodeling of the placenta reduces perfusion, leading to placental ischemia. Subsequently, the ischemic placenta releases antiangiogenic and pro-inflammatory factors, such as cytokines, reactive oxygen species, and the angiotensin II type 1 receptor autoantibody (AT1-AA), among others, into the maternal circulation. These factors cause widespread endothelial activation, upregulation of the endothelin system, and vasoconstriction. In turn, these changes affect the function of multiple organ systems including the kidneys, brain, liver, and heart. Despite extensive research into the pathophysiology of PE, the only treatment option remains early delivery of the baby and importantly, the placenta. While premature delivery is effective in ameliorating immediate risk to the mother, mounting evidence suggests that PE increases risk of cardiovascular disease later in life for both mother and baby. Notably, these women are at increased risk of hypertension, heart disease, and stroke, while offspring are at risk of obesity, hypertension, and neurological disease, among other complications, later in life. This article aims to discuss the current understanding of the diagnosis and pathophysiology of PE, as well as associated organ damage, maternal and fetal outcomes, and potential therapeutic avenues. © 2021 American Physiological Society. Compr Physiol 11:1315-1349, 2021.
Collapse
Affiliation(s)
- Bhavisha A. Bakrania
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Frank T. Spradley
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Surgery, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Heather A. Drummond
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Babbette LaMarca
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael J. Ryan
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Joey P. Granger
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
25
|
Maeda KJ, McClung DM, Showmaker KC, Warrington JP, Ryan MJ, Garrett MR, Sasser JM. Endothelial cell disruption drives increased blood-brain barrier permeability and cerebral edema in the Dahl SS/jr rat model of superimposed preeclampsia. Am J Physiol Heart Circ Physiol 2020; 320:H535-H548. [PMID: 33275518 DOI: 10.1152/ajpheart.00383.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Preeclampsia is characterized by increases in blood pressure and proteinuria in late pregnancy, and neurological symptoms can appear in the form of headaches, blurred vision, cerebral edema, and, in the most severe cases, seizures (eclampsia). The causes for these cerebral manifestations remain unknown, so the use of animal models that mimic preeclampsia is essential to understanding its pathogenesis. The Dahl salt-sensitive (Dahl SS/jr) rat model develops spontaneous preeclampsia superimposed on chronic hypertension; therefore, we hypothesized that the Dahl SS/jr rat would display cerebrovascular features similar to those seen in human preeclampsia. Furthermore, we predicted that this model would allow for the identification of mechanisms underlying these changes. The pregnant Dahl SS/jr rat displayed increased cerebral edema and blood-brain barrier disruption despite tighter control of cerebral blood flow autoregulation and vascular smooth muscle myogenic tone. Analysis of cerebral endothelial cell morphology revealed increased opening of tight junctions, basement membrane dissolution, and vesicle formation. RNAseq analysis identified that genes related to endothelial cell tight junctions and blood-brain barrier integrity were differentially expressed in cerebral vessels from pregnant Dahl SS/jr compared with healthy pregnant Sprague Dawley rats. Overall, our data reveal new insights into mechanisms involved in the cerebrovascular dysfunction of preeclampsia.NEW & NOTEWORTHY This study uses the Dahl SS/jr rat as a preclinical model of spontaneous superimposed preeclampsia to demonstrate uncoupling of cerebral vascular permeability and blood-brain barrier disruption from cerebral blood flow autoregulatory dysfunction and myogenic tone. Additionally, the data presented in this study lay the foundational framework on which future experiments assessing specific transcellular transport components such as individual transporter protein expression and components of the vesicular transport system (caveolae) can be built to help reveal a potential direct mechanistic insight into the causes of cerebrovascular complications during preeclamptic pregnancies.
Collapse
Affiliation(s)
- Kenji J Maeda
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Daniel M McClung
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kurt C Showmaker
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi
| | - Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer M Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
26
|
Duncan JW, Azubuike D, Booz GW, Fisher B, Williams JM, Fan F, Ibrahim T, LaMarca B, Cunningham MW. Angiotensin II type 1 receptor autoantibody blockade improves cerebral blood flow autoregulation and hypertension in a preclinical model of preeclampsia. Hypertens Pregnancy 2020; 39:451-460. [PMID: 33119997 DOI: 10.1080/10641955.2020.1833215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction:Women with preeclampsia (PE) and reduced uterine perfusion pressure (RUPP) pre-clinical rat model of PE have elevated angiotensin II type 1 receptor agonistic autoantibodies (AT1-AA) and cerebrovascular dysfunction. Methods:Sprague Dawley rats had RUPP surgery with/without AT1-AA inhibitor ('n7AAc'144 μg/day) osmotic minipumps. Mean arterial pressure (MAP), CBF autoregulation, blood brain barrier (BBB) permeability, cerebral edema, oxidative stress, and eNOS were assessed. Results:'n7AAc' improved MAP, restored CBF autoregulation, prevented cerebral edema, elevated oxidative stress, and increased phosphorylated eNOS protein in RUPP rats. Conclusion:Inhibiting the AT1-AA in placental ischemic rats prevents hypertension, cerebrovascular dysfunction, and improves cerebral metabolic function.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, MS, USA
| | - Daniel Azubuike
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Brandon Fisher
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA.,Department of Obstetrics and Gynecology, University of Mississippi Medical Center , Jackson, MS, USA
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, MS, USA
| |
Collapse
|
27
|
Wang S, Lv W, Zhang H, Liu Y, Li L, Jefferson JR, Guo Y, Li M, Gao W, Fang X, Paul IA, Rajkowska G, Shaffery JP, Mosley TH, Hu X, Liu R, Wang Y, Yu H, Roman RJ, Fan F. Aging exacerbates impairments of cerebral blood flow autoregulation and cognition in diabetic rats. GeroScience 2020; 42:1387-1410. [PMID: 32696219 DOI: 10.1007/s11357-020-00233-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a leading risk factor for aging-related dementia; however, the underlying mechanisms are not well understood. The present study, utilizing a non-obese T2DN diabetic model, demonstrates that the myogenic response of the middle cerebral artery (MCA) and parenchymal arteriole (PA) and autoregulation of cerebral blood flow (CBF) in the surface and deep cortex were impaired at both young and old ages. The impaired CBF autoregulation was more severe in old than young DM rats, and in the deep than the surface cortex. The myogenic tone of the MCA was enhanced at perfusion pressure in the range of 40-100 mmHg in young DM rats but was reduced at 140-180 mmHg in old DM rats. No change of the myogenic tone of the PA was observed in young DM rats, whereas it was significantly reduced at 30-60 mmHg in old DM rats. Old DM rats had enhanced blood-brain barrier (BBB) leakage and neurodegeneration, reduced vascular density, tight junction, and pericyte coverage on cerebral capillaries in the CA3 region in the hippocampus. Additionally, DM rats displayed impaired functional hyperemia and spatial learning and short- and long-term memory at both young and old ages. Old DM rats had impaired non-spatial short-term memory. These results revealed that impaired CBF autoregulation and enhanced BBB leakage plays an essential role in the pathogenesis of age- and diabetes-related dementia. These findings will lay the foundations for the discovery of anti-diabetic therapies targeting restoring CBF autoregulation to prevent the onset and progression of dementia in elderly DM.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.,Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Longyang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Man Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ian A Paul
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - James P Shaffery
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Thomas H Mosley
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.,Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Xinlin Hu
- Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Ruen Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yangang Wang
- Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
28
|
Prior exposure to placental ischemia causes increased salt sensitivity of blood pressure via vasopressin production and secretion in postpartum rats. J Hypertens 2020; 37:1657-1667. [PMID: 30950978 DOI: 10.1097/hjh.0000000000002091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Women with a history of preeclampsia exhibit increased salt sensitivity of blood pressure at postpartum, which might be responsible for their increased risk of future cardiovascular diseases. However, it is unclear whether preeclampsia can cause increased salt sensitivity at postpartum. Vasopressin may play a role in the pathogenesis of preeclampsia and salt-sensitive hypertension. Therefore, the aim of this study was to determine whether the exposure to preeclampsia, as elicited by placental ischemia, causes increased salt sensitivity at postpartum, and if so, whether vasopressin is involved in its process. METHODS AND RESULTS We used a reduced uterine perfusion pressure (RUPP) rat model of preeclampsia. Pregnant Sprague-Dawley rats were categorized into the following two groups: RUPP-operated and sham-operated (SHAM) control groups. A 1-week-long high-salt diet was initiated at 3 weeks postpartum. The high-salt diet-induced increase in mean arterial pressure was significantly greater in the RUPP group than in the SHAM group. In addition, the plasma levels of copeptin, a substitute for plasma vasopressin, increased and serum osmolality decreased in the RUPP group. Double immunostaining revealed that the expression of c-Fos, a marker of neural activity, in vasopressin-producing neurons and presympathetic neurons in the hypothalamic paraventricular nucleus was significantly elevated in the RUPP group. The oral administration of conivaptan, the dual V1a/V2 vasopressin receptor antagonist, during high-salt diet abolished the enhanced increase in mean arterial pressure in RUPP rats. CONCLUSION Prior exposure to placental ischemia causes increased salt sensitivity of blood pressure at postpartum probably due to enhanced vasopressin production and secretion.
Collapse
|
29
|
Interleukin-17 Reduces βENaC via MAPK Signaling in Vascular Smooth Muscle Cells. Int J Mol Sci 2020; 21:ijms21082953. [PMID: 32331392 PMCID: PMC7215799 DOI: 10.3390/ijms21082953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 11/23/2022] Open
Abstract
Degenerin proteins, such as the beta epithelial Na+ channel (βENaC), are essential in the intracellular signaling of pressure-induced constriction, an important vascular smooth muscle cell (VSMC) function. While certain cytokines reduce ENaC protein in epithelial tissue, it is unknown if interleukin-17 (IL-17), a potent pro-inflammatory cytokine, directly mediates changes in membrane-associated βENaC in VSMCs. Therefore, we tested the hypothesis that exposure to IL-17 reduces βENaC in VSMCs through canonical mitogen-activated protein kinase (MAPK) signaling pathways. We treated cultured rat VSMCs (A10 cell line) with IL-17 (1–100 ng/mL) for 15 min to 16 h and measured expression of βENaC, p38MAPK, c-jun kinase (JNK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). IL-17 reduced βENaC protein expression in a concentration-dependent fashion and increased phosphorylation of p38MAPK by 15 min and JNK by 8 h. NFκB was unaffected by IL-17 in VSMCs. IL-17 treatment reduced VSMC viability but had no effect on cell death. To determine the underlying signaling pathway involved in this response, VSMCs were treated before and during IL-17 exposure with p38MAPK or JNK inhibitors. We found that JNK blockade prevented IL-17-mediated βENaC protein suppression. These data demonstrate that the pro-inflammatory cytokine IL-17 regulates VSMC βENaC via canonical MAPK signaling pathways, raising the possibility that βENaC-mediated loss of VSMC function may occur in inflammatory disorders.
Collapse
|
30
|
Wang S, Zhang H, Liu Y, Li L, Guo Y, Jiao F, Fang X, Jefferson JR, Li M, Gao W, Gonzalez-Fernandez E, Maranon RO, Pabbidi MR, Liu R, Alexander BT, Roman RJ, Fan F. Sex differences in the structure and function of rat middle cerebral arteries. Am J Physiol Heart Circ Physiol 2020; 318:H1219-H1232. [PMID: 32216612 PMCID: PMC7346534 DOI: 10.1152/ajpheart.00722.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidemiological studies demonstrate that there are sex differences in the incidence, prevalence, and outcomes of cerebrovascular disease (CVD). The present study compared the structure and composition of the middle cerebral artery (MCA), neurovascular coupling, and cerebrovascular function and cognition in young Sprague-Dawley (SD) rats. Wall thickness and the inner diameter of the MCA were smaller in females than males. Female MCA exhibited less vascular smooth muscle cells (VSMCs), diminished contractile capability, and more collagen in the media, and a thicker internal elastic lamina with fewer fenestrae compared with males. Female MCA had elevated myogenic tone, lower distensibility, and higher wall stress. The stress/strain curves shifted to the left in female vessels compared with males. The MCA of females failed to constrict compared with a decrease of 15.5 ± 1.9% in males when perfusion pressure was increased from 40 to 180 mmHg. Cerebral blood flow (CBF) rose by 57.4 ± 4.4 and 30.1 ± 3.1% in females and males, respectively, when perfusion pressure increased from 100 to 180 mmHg. The removal of endothelia did not alter the myogenic response in both sexes. Functional hyperemia responses to whisker-barrel stimulation and cognition examined with an eight-arm water maze were similar in both sexes. These results demonstrate that there are intrinsic structural differences in the MCA between sexes, which are associated with diminished myogenic response and CBF autoregulation in females. The structural differences do not alter neurovascular coupling and cognition at a young age; however, they might play a role in the development of CVD after menopause. NEW & NOTEWORTHY Using perfusion fixation of the middle cerebral artery (MCA) in calcium-free solution at physiological pressure and systematically randomly sampling the sections prepared from the same M2 segments of MCA, we found that there are structural differences that are associated with altered cerebral blood flow (CBF) autoregulation but not neurovascular coupling and cognition in young, healthy Sprague-Dawley (SD) rats. Understanding the intrinsic differences in cerebrovascular structure and function in males and females is essential to develop new pharmaceutical treatments for cerebrovascular disease (CVD).
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Longyang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Feng Jiao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Neurosurgery, Peking University People's Hospital, Beijing, China
| | - Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Man Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ezekiel Gonzalez-Fernandez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rodrigo O Maranon
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ruen Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing, China
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
31
|
Duncan JW, Younes ST, Hildebrandt E, Ryan MJ, Granger JP, Drummond HA. Tumor necrosis factor-α impairs cerebral blood flow in pregnant rats: role of vascular β-epithelial Na + channel. Am J Physiol Heart Circ Physiol 2020; 318:H1018-H1027. [PMID: 32167780 DOI: 10.1152/ajpheart.00744.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Preeclampsia is a pregnancy-related disorder characterized by hypertension, vascular dysfunction and an increase in circulating inflammatory factors including the cytokine, tumor necrosis factor-α (TNF-α). Studies have shown that placental ischemia is associated with 1) increased circulating TNF-α, 2) attenuated pressure-induced cerebral vascular tone, and 3) suppression of β-epithelial Na+ channel (βENaC) protein in cerebral vessels. In addition to its role in epithelial Na+ and water transport, βENaC is an essential signaling element in transduction of pressure-induced (aka "myogenic") constriction, a critical mechanism of blood flow autoregulation. While cytokines inhibit expression of certain ENaC proteins in epithelial tissue, it is unknown if the increased circulating TNF-α associated with placental ischemia mediates the loss of cerebrovascular βENaC and cerebral blood flow regulation. Therefore, the purpose of this study was to test the hypothesis that increasing plasma TNF-α in normal pregnant rats reduces cerebrovascular βENaC expression and impairs cerebral blood flow (CBF) regulation. In vivo TNF-α infusion (200 ng/day, 5 days) inhibited cerebrovascular expression of βENaC and impaired CBF regulation in pregnant rats. To determine the direct effects of TNF-α and underlying pathways mediating vascular smooth muscle cell βENaC reduction, we exposed cultured VSMCs (A10 cell line) to TNF-α (1-100 ng/mL) for 16-24 h. TNF-α reduced βENaC protein expression in a concentration-dependent fashion from 0.1 to 100 ng/mL, without affecting cell death. To assess the role of canonical MAPK signaling in this response, VSMCs were treated with p38MAPK or c-Jun kinase (JNK) inhibitors in the presence of TNF-α. We found that both p38MAPK and JNK blockade prevented TNF-α-mediated βENaC protein suppression. These data provide evidence that disorders associated with increased circulating TNF-α could lead to impaired cerebrovascular regulation, possibly due to reduced βENaC-mediated vascular function.NEW & NOTEWORTHY This manuscript identifies TNF-α as a possible placental-derived cytokine that could be involved in declining cerebrovascular health observed in preeclampsia. We found that infusion of TNF-α during pregnancy impaired cerebral blood flow control in rats at high arterial pressures. We further discovered that cerebrovascular β-epithelial sodium channel (βENaC) protein, a degenerin protein involved in mechanotransduction, was reduced by TNF-α in pregnant rats, indicating a potential link between impaired blood flow and this myogenic player. We next examined this effect in vitro using a rat vascular smooth muscle cell line. TNF-α reduced βENaC through canonical MAPK-signaling pathways and was not dependent on cell death. This study demonstrates the pejorative effects of TNF-α on cerebrovascular function during pregnancy and warrants future investigations to study the role of cytokines on vascular function during pregnancy.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Subhi Talal Younes
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Emily Hildebrandt
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Heather A Drummond
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
32
|
Spradley FT. Targeting inflammation to reduce seizure severity in an experimental model of eclampsia. Hypertens Res 2020; 43:350-353. [PMID: 31932641 DOI: 10.1038/s41440-019-0317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Frank T Spradley
- Department of Surgery, The University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
33
|
Murali S, Miller K, McDermott M. Preeclampsia, eclampsia, and posterior reversible encephalopathy syndrome. HANDBOOK OF CLINICAL NEUROLOGY 2020; 172:63-77. [PMID: 32768095 DOI: 10.1016/b978-0-444-64240-0.00004-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Preeclampsia is a disorder of pregnancy associated with gestational hypertension and end-organ dysfunction. Patients with eclampsia, by definition, have seizures as part of the clinical syndrome. However, patients with preeclampsia can also have other neurologic symptoms and deficits. Both disorders can be associated with radiographic abnormalities similar to that of posterior reversible encephalopathy syndrome, suggesting a common pathophysiology or unified clinical spectrum of disorders. This chapter reviews the pathophysiology, clinical presentation, diagnostic findings, and prognosis of patients with neurologic complications associated with preeclampsia and eclampsia.
Collapse
Affiliation(s)
- Sadhana Murali
- Department of Neurology, University of Michigan Stroke Program, Ann Arbor, MI, United States
| | - Kristin Miller
- Department of Neurology, University of Illinois at Chicago, Stroke Program, Chicago, IL, United States
| | - Mollie McDermott
- Department of Neurology, University of Michigan Stroke Program, Ann Arbor, MI, United States.
| |
Collapse
|
34
|
Warrington JP, Fan F, Duncan J, Cunningham MW, LaMarca BB, Dechend R, Wallukat G, Roman RJ, Drummond HA, Granger JP, Ryan MJ. The angiotensin II type I receptor contributes to impaired cerebral blood flow autoregulation caused by placental ischemia in pregnant rats. Biol Sex Differ 2019; 10:58. [PMID: 31829239 PMCID: PMC6907203 DOI: 10.1186/s13293-019-0275-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/29/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Placental ischemia and hypertension, characteristic features of preeclampsia, are associated with impaired cerebral blood flow (CBF) autoregulation and cerebral edema. However, the factors that contribute to these cerebral abnormalities are not clear. Several lines of evidence suggest that angiotensin II can impact cerebrovascular function; however, the role of the renin angiotensin system in cerebrovascular function during placental ischemia has not been examined. We tested whether the angiotensin type 1 (AT1) receptor contributes to impaired CBF autoregulation in pregnant rats with placental ischemia caused by surgically reducing uterine perfusion pressure. METHODS Placental ischemic or sham operated rats were treated with vehicle or losartan from gestational day (GD) 14 to 19 in the drinking water. On GD 19, we assessed CBF autoregulation in anesthetized rats using laser Doppler flowmetry. RESULTS Placental ischemic rats had impaired CBF autoregulation that was attenuated by treatment with losartan. In addition, we examined whether an agonistic autoantibody to the AT1 receptor (AT1-AA), reported to be present in preeclamptic women, contributes to impaired CBF autoregulation. Purified rat AT1-AA or vehicle was infused into pregnant rats from GD 12 to 19 via mini-osmotic pumps after which CBF autoregulation was assessed. AT1-AA infusion impaired CBF autoregulation but did not affect brain water content. CONCLUSIONS These results suggest that the impaired CBF autoregulation associated with placental ischemia is due, at least in part, to activation of the AT1 receptor and that the RAS may interact with other placental factors to promote cerebrovascular changes common to preeclampsia.
Collapse
Affiliation(s)
- Junie P Warrington
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Jeremy Duncan
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Mark W Cunningham
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Babette B LaMarca
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Ralf Dechend
- Experimental and Clinical Research Center and Max-Delbrück Center for Molecular Medicine, and HELIOS Clinic Berlin, Berlin, Germany
| | - Gerd Wallukat
- Experimental and Clinical Research Center and Max-Delbrück Center for Molecular Medicine, and HELIOS Clinic Berlin, Berlin, Germany
| | - Richard J Roman
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Heather A Drummond
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Joey P Granger
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Michael J Ryan
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
35
|
Miller KB, Miller VM, Harvey RE, Ranadive SM, Joyner MJ, Barnes JN. Augmented cerebral blood velocity in response to isometric handgrip exercise in women with a history of preeclampsia. Am J Physiol Regul Integr Comp Physiol 2019; 317:R834-R839. [PMID: 31663771 DOI: 10.1152/ajpregu.00280.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy described as a condition of excessive sympathoexcitation. PE places a woman at increased risk for lifelong hypertension and cognitive impairment. Cerebral blood velocity is blunted in response to a vasoactive stimulus in women with a history of PE. This study investigated how a sympathoexcitatory stimulus affects cerebral blood velocity in women with a history of PE. Middle cerebral artery blood velocity (MCAv) and beat-to-beat mean arterial blood pressure (MAP) were measured in postmenopausal women with a history of PE (n = 21; age = 59 ± 5 yr) and a history of a normotensive pregnancy (NP; n = 27; age = 58 ± 4 yr), at baseline, during isometric handgrip to fatigue (IHG) followed by postexercise ischemia (PEI), and a recovery period (REC). Baseline MAP and MAP responses to IHG and PEI did not differ between groups. MCAv at baseline and throughout the stimulus was lower in PE women compared with NP women (P < 0.05 for all). MCAv increased during IHG in both groups (P < 0.05). This increase in MCAv was greater in PE compared with NP women during IHG and REC (IHG: PE 13 ± 2% vs. NP 9 ± 2%; REC: PE 3 ± 2% vs. NP -2 ± 2%; P < 0.05 for both). Thus, a history of PE is associated with low baseline cerebral blood velocity but an augmented response to a sympathoexcitatory stimulus. These changes in cerebral blood flow regulation may lead to an increased risk for cognitive impairment in women with a history of PE.
Collapse
Affiliation(s)
- Kathleen B Miller
- Bruno Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Virginia M Miller
- Departments of Surgery, and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Ronée E Harvey
- Department of Internal Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Sushant M Ranadive
- Department of Kinesiology, University of Maryland, College Park, Maryland
| | | | - Jill N Barnes
- Bruno Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
36
|
Cerebral Blood Flow Regulation in Pregnancy, Hypertension, and Hypertensive Disorders of Pregnancy. Brain Sci 2019; 9:brainsci9090224. [PMID: 31487961 PMCID: PMC6769869 DOI: 10.3390/brainsci9090224] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 01/12/2023] Open
Abstract
The regulation of cerebral blood flow (CBF) allows for the metabolic demands of the brain to be met and for normal brain function including cognition (learning and memory). Regulation of CBF ensures relatively constant blood flow to the brain despite changes in systemic blood pressure, protecting the fragile micro-vessels from damage. CBF regulation is altered in pregnancy and is further altered by hypertension and hypertensive disorders of pregnancy including preeclampsia. The mechanisms contributing to changes in CBF in normal pregnancy, hypertension, and preeclampsia have not been fully elucidated. This review summarizes what is known about changes in CBF regulation during pregnancy, hypertension, and preeclampsia.
Collapse
|
37
|
Giambrone AB, Logue OC, Shao Q, Bidwell GL, Warrington JP. Perinatal Micro-Bleeds and Neuroinflammation in E19 Rat Fetuses Exposed to Utero-Placental Ischemia. Int J Mol Sci 2019; 20:ijms20164051. [PMID: 31434191 PMCID: PMC6720786 DOI: 10.3390/ijms20164051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/15/2019] [Accepted: 08/18/2019] [Indexed: 12/24/2022] Open
Abstract
Offspring of preeclampsia patients have an increased risk of developing neurological deficits and cognitive impairment. While low placental perfusion, common in preeclampsia and growth restriction, has been linked to neurological deficits, a causative link is not fully established. The goal of this study was to test the hypothesis that placental ischemia induces neuroinflammation and micro-hemorrhages in utero. Timed-pregnant Sprague Dawley rats were weight-matched for sham surgery (abdominal incision only) or induced placental ischemia (surgical reduction of utero-placental perfusion (RUPP)); n = 5/group on gestational day 14. Fetal brains (n = 1–2/dam/endpoint) were collected at embryonic day (E19). Placental ischemia resulted in fewer live fetuses, increased fetal demise, increased hematocrit, and no difference in brain water content in exposed fetuses. Additionally, increased cerebral micro-bleeds (identified with H&E staining), pro-inflammatory cytokines: IL-1β, IL-6, and IL-18, eotaxin (CCL11), LIX (CXCL5), and MIP-2 (CXCL2) were observed in RUPP-exposed fetuses. Microglial density in the sub-ventricular zone decreased in RUPP-exposed fetuses, with no change in cortical thickness. Our findings support the hypothesis that exposure to placental ischemia contributes to microvascular dysfunction (increased micro-bleeds), fetal brain inflammation, and reduced microglial density in proliferative brain areas. Future studies will determine whether in utero abnormalities contribute to long-term behavioral deficits in preeclampsia offspring through impaired neurogenesis regulation.
Collapse
Affiliation(s)
- Ashtin B Giambrone
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Omar C Logue
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Qingmei Shao
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
38
|
Bergman L, Torres-Vergara P, Penny J, Wikström J, Nelander M, Leon J, Tolcher M, Roberts JM, Wikström AK, Escudero C. Investigating Maternal Brain Alterations in Preeclampsia: the Need for a Multidisciplinary Effort. Curr Hypertens Rep 2019; 21:72. [PMID: 31375930 DOI: 10.1007/s11906-019-0977-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To provide insight into the mechanisms underlying cerebral pathophysiology and to highlight possible methods for evaluation, screening, and surveillance of cerebral complications in preeclampsia. RECENT FINDINGS The pathophysiology of eclampsia remains enigmatic. Animal studies show that the cerebral circulation in pregnancy and preeclampsia might be affected with increased permeability over the blood-brain barrier and altered cerebral blood flow due to impaired cerebral autoregulation. The increased blood pressure cannot be the only underlying cause of eclampsia and cerebral edema, since some cases of eclampsia arise without simultaneous hypertension. Findings from animal studies need to be confirmed in human tissues. Evaluation of brain alterations in preeclampsia and eclampsia is challenging and demands a multidisciplinary collaboration, since no single method can accurately and fully describe how preeclampsia affects the brain. Cerebral complications of preeclampsia are significant factors in maternal morbidity and mortality worldwide. No single method can accurately describe the full picture of how preeclampsia affects the brain vasculature and parenchyma. We recommend an international and multidisciplinary effort not only to overcome the issue of limited sample availability but also to optimize the quality of research.
Collapse
Affiliation(s)
- Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
- Center for Clinical Research Dalarna, Falun, Uppsala, Sweden.
| | - Pablo Torres-Vergara
- Pharmacy Department, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Johan Wikström
- Department of Radiology, Uppsala University, Uppsala, Sweden
| | - Maria Nelander
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Jose Leon
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis, (LFV-GIANT), Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Mary Tolcher
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - James M Roberts
- Magee Womens Research Institute, Dept of Obstetrics Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis, (LFV-GIANT), Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile.
| |
Collapse
|
39
|
Affiliation(s)
- Eliza C Miller
- From the Department of Neurology, Division of Stroke and Cerebrovascular Disease, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW To review and summarize what is known about cerebrovascular derangements during preeclampsia. RECENT FINDINGS Preeclampsia is a devastating disorder of pregnancy with no known cure. Little is known about the pathophysiological mechanisms which lead to the symptoms of the disorder, particularly with regard to individual vascular beds such as the cerebral circulation. Studies suggest that the cerebrovascular dysfunction characteristic of the preeclampsia syndrome is characterized by alterations in cerebral blood flow autoregulation and opening of the blood-brain barrier. Mechanistic studies demonstrate that the same circulating factors implicated in the pathophysiology of other vascular beds may be operative in the cerebral circulation as well. However, significant knowledge gaps still exist, highlighting the need for more intense research in this field. Little is known about cerebrovascular dysfunction during preeclampsia, and detailed mechanistic studies are needed to identify the molecular pathways involved, the interactions thereof, and how those pathways lead to clinical disease.
Collapse
Affiliation(s)
- Subhi Talal Younes
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216-4505, USA
| | - Michael J Ryan
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216-4505, USA.
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
41
|
Wallace K, Bean C, Bowles T, Spencer SK, Randle W, Kyle PB, Shaffery J. Hypertension, Anxiety, and Blood-Brain Barrier Permeability Are Increased in Postpartum Severe Preeclampsia/Hemolysis, Elevated Liver Enzymes, and Low Platelet Count Syndrome Rats. Hypertension 2019; 72:946-954. [PMID: 30354708 DOI: 10.1161/hypertensionaha.118.11770] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension and inflammation during pregnancy are suggested to contribute to the development of postpartum depression and anxiety. Using a rat model of severe preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, which displays both hypertension and inflammation during pregnancy, we evaluated whether rats were prone to develop depression or anxiety in the postpartum period. On gestational day 12, miniosmotic pumps infusing sFlt-1 (soluble fms-like tyrosine kinase-1) and sEng (soluble endoglin) were placed into rats, a subset of these rats was infused with 2 mg/kg of Orencia (abatacept) the following day to determine whether immune suppression via T-cell depletion prevented any changes in maternal depression or anxiety-like behavior. All rats, including normal pregnant (NP) controls, delivered between gestational days 21 and 22. Postpartum severe preeclamptic rats buried significantly more marbles compared with NP rats ( P=0.002) and Orencia-treated rats ( P=0.05). Severe preeclamptic rats spent significantly more time in closed arms of the elevated plus maze compared with NP rats ( P=0.009) and Orencia-treated rats ( P=0.05). Severe preeclamptic rats were hypertensive compared with NP ( P=0.03) and Orencia-treated rats ( P=0.01). Finally, severe preeclamptic rats had increased blood-brain barrier permeability compared with NP rats ( P=0.03), which was reversed in Orencia-treated rats ( P=0.008). These results suggest that severe preeclampsia/hemolysis, elevated liver enzymes, and low platelet count syndrome during pregnancy contributes to an increase in anxiety-like behavior, blood-brain barrier permeability, and hypertension in the postpartum. The current results suggest that T-cell suppression during pregnancy can also help prevent chronic hypertension and increased anxiety in the postpartum period.
Collapse
Affiliation(s)
- Kedra Wallace
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Cynthia Bean
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Teylor Bowles
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Shauna-Kay Spencer
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Wisdom Randle
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Patrick B Kyle
- Department of Pathology (P.B.K.), University of Mississippi Medical Center, Jackson
| | - James Shaffery
- Department of Psychiatry, Center of Psychiatric Neuroscience (J.S.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
42
|
Peripheral Anti-Angiogenic Imbalance during Pregnancy Impairs Myogenic Tone and Increases Cerebral Edema in a Rodent Model of HELLP Syndrome. Brain Sci 2018; 8:brainsci8120216. [PMID: 30563221 PMCID: PMC6316026 DOI: 10.3390/brainsci8120216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 01/14/2023] Open
Abstract
Using an animal model of hemolysis elevated liver enzymes low platelets (HELLP) that has systemic inflammation and neuroinflammation we wanted to determine if blood brain barrier (BBB) permeability, cerebral edema, vascular tone, and occludin expression were altered in pregnant rats. Anti-angiogenic proteins sFlt-1 and sEng (4.7 and 7 µg/kg/day, respectively) were chronically infused into normal pregnant (NP) rats beginning on gestational day 12 via a mini-osmotic pump. On gestational day 19, blood pressure was measured via a carotid catheter and brains were collected. BBB permeability was assessed in select brain regions from rats infused with 0.5 mg/mL Texas Red Dextran and phenylephrine. Occludin, sFlt-1, and sEng were analyzed via western blot or ELISA. Infusion of sFlt-1 and sEng into NP rats increased hemolysis and liver enzymes, and decreased platelets and led to hypertension. HELLP rats had significant impairment in the myogenic response and increased BBB permeability in the posterior cortex and brainstem. Brain water content in the posterior cortex was increased and sEng protein expression in the brainstem was significantly increased in HELLP rats. The results from this study suggest that a peripheral anti-angiogenic imbalance during pregnancy is associated with decreased myogenic tone, vasogenic edema, and an increase in BBB permeability, but not anti-angiogenic imbalance in the brain.
Collapse
|
43
|
The Effects of Lipopolysaccharide on the Disrupted Blood-Brain Barrier in a Rat Model of Preeclampsia. J Stroke Cerebrovasc Dis 2018; 27:3411-3418. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 01/26/2023] Open
|
44
|
Torres-Vergara P, Escudero C, Penny J. Drug Transport at the Brain and Endothelial Dysfunction in Preeclampsia: Implications and Perspectives. Front Physiol 2018; 9:1502. [PMID: 30459636 PMCID: PMC6232255 DOI: 10.3389/fphys.2018.01502] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022] Open
Abstract
Transport of drugs across biological barriers has been a subject of study for decades. The discovery and characterization of proteins that confer the barrier properties of endothelia and epithelia, including tight junction proteins and membrane transporters belonging to the ATP-binding cassette (ABC) and Solute Carrier (SLC) families, represented a significant step forward into understanding the mechanisms that govern drug disposition. Subsequently, numerous studies, including both pre-clinical approaches and clinical investigations, have been carried out to determine the influence of physiological and pathological states on drug disposition. Importantly, there has been increasing interest in gaining a better understanding of drug disposition during pregnancy, since epidemiological and clinical studies have demonstrated that the use of medications by pregnant women is significant and this condition embodies a series of significant anatomical and physiological modifications, particularly at excretory organs and barrier sites (e.g., placenta, breast) expressing transporter proteins which influence pharmacokinetics. Currently, most of the research in this field has focused on the expression profiling of transporter proteins in trophoblasts and endothelial cells of the placenta, regulation of drug-resistance mechanisms in disease states and pharmacokinetic studies. However, little attention has been placed on the influence that the cerebrovascular dysfunction present in pregnancy-related disorders, such as preeclampsia, might exert on drug disposition in the mother’s brain. This issue is particularly important since recent findings have demonstrated that preeclamptic women suffer from long-term alterations in the integrity of the blood-brain barrier (BBB). In this review we aim to analyze the available evidence regarding the influence of pregnancy on the expression of transporters and TJ proteins in brain endothelial cells, as well the mechanisms that govern the pathophysiological alterations in the BBB of women who experience preeclampsia. Future research efforts should be focused not only on achieving a better understanding of the influence of preeclampsia-associated endothelial dysfunction on drug disposition, but also in optimizing the pharmacological treatments of women suffering pregnancy-related disorders, its comorbidities and to develop new therapies aiming to restore the integrity of the BBB.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Concepción, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Red Iberoamericana de Alteraciones Vasculares Asociadas a Trastornos del Embarazo (RIVA-TREM), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
45
|
Xie YL, Zhang B, Jing L. MiR-125b blocks Bax/Cytochrome C/Caspase-3 apoptotic signaling pathway in rat models of cerebral ischemia-reperfusion injury by targeting p53. Neurol Res 2018; 40:828-837. [PMID: 29956588 DOI: 10.1080/01616412.2018.1488654] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To explore the potential effect of miR-125b on p53-mediated regulation of Bax/Cytochrome C/Caspase-3 apoptotic signaling pathway in rats with cerebral ischemia-reperfusion (CIR) injury. METHODS Sprague-Dawley (SD) rats were used to conduct CIR injury and injected with miR-125b mimic/inhibitor or p53 inhibitor (Pifithrin-α, PFT-α). Dual-luciferase reporter gene assay was used to analyze the targeting relationship between miR-125b and p53. Longa scoring and Triphenyl tetrazolinm chloride (TTC) staining were used to test the neurologic function and determine infarct size, respectively. Hematoxylin-eosin (HE) and Nissl's stainings were conducted to observe the morphology of cortical neurons. Neuronal nuclei (NeuN) expression was detected by immunohistochemical staining. QRT-PCR was performed to detect the expressions of miR-125b and p53. TUNEL staining and Western blotting was used to determine neuronal apoptosis and expressions of Bax/Cytochrome C/Caspase-3 signaling pathway-related proteins, respectively. RESULTS Our results showed that miR-125b could directly target p53. As observed, overexpression of miR-125b could obviously reduce the neurological score, infarct size, and brain water content after CIR in rats, which also improved the morphology of cortical neurons, increased the number of neurons, reduced neuronal apoptosis, and inhibited the expressions of Bax/Cytochrome C/Caspase-3 pathway. Moreover,the similar results were observed in rats with CIR after injected with PFT-α. But no significant differences in each index were found in CIR group and CIR + anti-miR-125b + PFT-α group. CONCLUSION MiR-125b exerts protective effects on CIR injury through inhibition of Bax/Cytochrome C/Caspase-3signaling pathway via targeting p53, which is likely to be a promising treatment for CIR. ABBREVIATIONS 3'-UTR: 3-untranslated region; CIR: cerebral ischemia-reperfusion; CIS: cerebral ischemic stroke; PFT-α: Pifithrin-α; PVDF: polyvinylidene fluoride; SD: Sprague-Dawley; TBST: tris buffered saline with tween. TTC staining: Triphenyl tetrazolinm chloride staining; TUNEL: Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling.
Collapse
Affiliation(s)
- Yun-Liang Xie
- a Medical Department , The Affiliated Hospital of Bei Hua University , Jilin , China
| | - Bo Zhang
- b Health Care Department , The Affiliated Hospital of Bei Hua University , Jilin , China
| | - Ling Jing
- c College of Pharmacy , Jilin University , Changchun , China
| |
Collapse
|
46
|
Clayton AM, Shao Q, Paauw ND, Giambrone AB, Granger JP, Warrington JP. Postpartum increases in cerebral edema and inflammation in response to placental ischemia during pregnancy. Brain Behav Immun 2018; 70:376-389. [PMID: 29588233 PMCID: PMC5953832 DOI: 10.1016/j.bbi.2018.03.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/15/2018] [Accepted: 03/23/2018] [Indexed: 01/22/2023] Open
Abstract
Reduced placental blood flow results in placental ischemia, an initiating event in the pathophysiology of preeclampsia, a hypertensive pregnancy disorder. While studies show increased mortality risk from Alzheimer's disease, stroke, and cerebrovascular complications in women with a history of preeclampsia, the underlying mechanisms are unknown. During pregnancy, placental ischemia, induced by reducing uterine perfusion pressure (RUPP), leads to cerebral edema and increased blood-brain barrier (BBB) permeability; however whether these complications persist after delivery is not known. Therefore, we tested the hypothesis that placental ischemia contributes to postpartum cerebral edema and neuroinflammation. On gestational day 14, time-pregnant Sprague Dawley rats underwent Sham (n = 10) or RUPP (n = 9) surgery and brain tissue collected 2 months post-delivery. Water content increased in posterior cortex but not hippocampus, striatum, or anterior cerebrum following RUPP. Using a rat cytokine multi-plex kit, posterior cortical IL-17, IL-1α, IL-1β, Leptin, and MIP2 increased while hippocampal IL-4, IL-12(p70) and RANTES increased and IL-18 decreased following RUPP. Western blot analysis showed no changes in astrocyte marker, Glial Fibrillary Acidic Protein (GFAP); however, the microglia marker, ionized calcium binding adaptor molecule (Iba1) tended to increase in hippocampus of RUPP-exposed rats. Immunofluorescence staining revealed reduced number of posterior cortical microglia but increased activated (Type 4) microglia in RUPP. Astrocyte number increased in both regions but area covered by astrocytes increased only in posterior cortex following RUPP. BBB-associated proteins, Claudin-1, Aquaporin-4, and zonular occludens-1 expression were unaltered; however, posterior cortical occludin decreased. These results suggest that 2 months postpartum, neuroinflammation, along with decreased occludin expression, may partly explain posterior cortical edema in rats with history of placental ischemia.
Collapse
Affiliation(s)
| | | | - Nina D. Paauw
- Department of Obstetrics, Wilhelmina Children’s Hospital
Birth Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Joey P. Granger
- Department of Physiology & Biophysics, University of
Mississippi Medical Center, Jackson, MS 39216
| | | |
Collapse
|
47
|
Giambrone AB, Warrington JP. The rat model of placental ischemia as a model of postpartum posterior cortical atrophy? Neural Regen Res 2018; 13:2094-2095. [PMID: 30323132 PMCID: PMC6199940 DOI: 10.4103/1673-5374.241454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ashtin B Giambrone
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
48
|
Miller EC, Sundheim KM, Willey JZ, Boehme AK, Agalliu D, Marshall RS. The Impact of Pregnancy on Hemorrhagic Stroke in Young Women. Cerebrovasc Dis 2018; 46:10-15. [PMID: 29982254 PMCID: PMC6158089 DOI: 10.1159/000490803] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Pregnancy is a sex-specific risk factor for causing hemorrhagic stroke (HS) in young adults. Unique physiological characteristics during pregnancy may alter the relative risk for HS in pregnant/postpartum (PP) women compared to HS in other young women. We compared patient characteristics and HS subtypes between young non-pregnant and PP women. METHODS We reviewed the medical records of all women 18-45 years old admitted to our center with HS from October 15, 2008 through March 31, 2015, and compared patient characteristics and stroke mechanisms using logistic regression. RESULTS Of the 130 young women with HS during the study period, 111 were non-PP women, and 19 PP women. PP women had lower proportions of vascular risk factors such as hypertension, prior stroke, and smoking, and a higher proportion of migraine (36.8 vs. 14.4%, p = 0.01). After adjusting for hypertension, smoking, migraine, prior stroke and prior myocardial infarction, PP women had lower odds of having an underlying vascular lesion (OR 0.14, 95% CI 0.04-0.44, p = 0.0009) and a higher proportion of the reversible cerebral vasoconstriction syndrome (RCVS) as cause of their HS. CONCLUSIONS Women with pregnancy-associated HS had fewer cerebrovascular risk factors, lower odds of having -underlying vascular lesions, and higher proportion of -migraine and RCVS compared with similar-aged non--pregnant women. Pregnancy-associated HS appears to represent a unique pathophysiological process, requiring targeted study.
Collapse
Affiliation(s)
- Eliza C. Miller
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kathryn M. Sundheim
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Joshua Z. Willey
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Amelia K. Boehme
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA.,Departments of Pathology and Cell Biology and Pharmacology, Columbia University Medical Center, New York, NY, 10032, USA.,Columbia Neuroscience Translational Initiative Center, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, 10032, USA
| | - Randolph S. Marshall
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| |
Collapse
|
49
|
Preeclampsia and the brain: neural control of cardiovascular changes during pregnancy and neurological outcomes of preeclampsia. Clin Sci (Lond) 2017; 130:1417-34. [PMID: 27389588 DOI: 10.1042/cs20160108] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/13/2016] [Indexed: 02/07/2023]
Abstract
Preeclampsia (PE) is a form of gestational hypertension that complicates ∼5% of pregnancies worldwide. Over 70% of the fatal cases of PE are attributed to cerebral oedema, intracranial haemorrhage and eclampsia. The aetiology of PE originates from abnormal remodelling of the maternal spiral arteries, creating an ischaemic placenta that releases factors that drive the pathophysiology. An initial neurological outcome of PE is the absence of the autonomically regulated cardiovascular adaptations to pregnancy. PE patients exhibit sympathetic overactivation, in comparison with both normotensive pregnant and hypertensive non-pregnant females. Moreover, PE diminishes baroreceptor reflex sensitivity (BRS) beyond that observed in healthy pregnancy. The absence of the cardiovascular adaptations to pregnancy, combined with sympathovagal imbalance and a blunted BRS leads to life-threatening neurological outcomes. Behaviourally, the increased incidences of maternal depression, anxiety and post-traumatic stress disorder (PTSD) in PE are correlated to low fetal birth weight, intrauterine growth restriction (IUGR) and premature birth. This review addresses these neurological consequences of PE that present in the gravid female both during and after the index pregnancy.
Collapse
|
50
|
Siepmann T, Boardman H, Bilderbeck A, Griffanti L, Kenworthy Y, Zwager C, McKean D, Francis J, Neubauer S, Yu GZ, Lewandowski AJ, Sverrisdottir YB, Leeson P. Long-term cerebral white and gray matter changes after preeclampsia. Neurology 2017; 88:1256-1264. [PMID: 28235810 PMCID: PMC5373775 DOI: 10.1212/wnl.0000000000003765] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/22/2016] [Indexed: 01/03/2023] Open
Abstract
Objective: To determine whether changes in cerebral structure are present after preeclampsia that may explain increased cerebrovascular risk in these women. Methods: We conducted a case control study in women between 5 and 15 years after either a preeclamptic or normotensive pregnancy. Brain MRI was performed. Analysis of white matter structure was undertaken using voxel-based segmentation of fluid-attenuation inversion recovery sequences to assess white matter lesion volume and diffusion tensor imaging to measure microstructural integrity. Voxel-based analysis of gray matter volumes was performed with adjustment for skull size. Results: Thirty-four previously preeclamptic women (aged 42.8 ± 5.1 years) and 49 controls were included. Previously preeclamptic women had reduced cortical gray matter volume (523.2 ± 30.1 vs 544.4 ± 44.7 mL, p < 0.05) and, although both groups displayed white matter lesions, changes were more extensive in previously preeclamptic women. They displayed increased temporal lobe white matter disease (lesion volume: 23.2 ± 24.9 vs 10.9 ± 15.0 μL, p < 0.05) and altered microstructural integrity (radial diffusivity: 538 ± 19 vs 526 ± 18 × 10−6 mm2/s, p < 0.01), which also extended to occipital and parietal lobes. The degree of temporal lobe white matter change in previously preeclamptic women was independent of their current cardiovascular risk profile (p < 0.05) and increased with time from index pregnancy (p < 0.05). Conclusion: A history of preeclampsia is associated with temporal lobe white matter changes and reduced cortical volume in young women, which is out of proportion to their classic cardiovascular risk profile. The severity of changes is proportional to time since pregnancy, which would be consistent with continued accumulation of damage after pregnancy.
Collapse
Affiliation(s)
- Timo Siepmann
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Henry Boardman
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Amy Bilderbeck
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ludovica Griffanti
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Yvonne Kenworthy
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Charlotte Zwager
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - David McKean
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jane Francis
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Neubauer
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Grace Z Yu
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Adam J Lewandowski
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Yrsa Bergmann Sverrisdottir
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Paul Leeson
- From the Radcliffe Department of Medicine (T.S., H.B., Y.K., C.Z., J.F., S.N., A.J.L., P.L.), Department of Psychiatry (A.B.), Nuffield Department of Clinical Neurosciences (L.G.), Nuffield Department of Surgical Sciences (Y.B.S.), and Department of Cardiology (G.Z.Y.), University of Oxford; Department of Radiology (D.M.), Stoke Mandeville Hospital, Aylesbury, UK; and Department of Neurology (T.S.), University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|