1
|
Wang Y, Wang H, Lu H, Ma J, Wu W, Wang Y, Ma B, Zhu H, Hu R. Renal glomerular and tubular injury in the offspring of the preeclampsia-like syndrome. Sci Rep 2025; 15:915. [PMID: 39762506 PMCID: PMC11704207 DOI: 10.1038/s41598-025-85258-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Preeclampsia (PE) is a prevalent and severe pregnancy complication that significantly impacts maternal and perinatal health. Epidemiological studies and animal experiments have demonstrated that PE adversely affects the cardiovascular and nervous systems of offspring, increasing their risk of hypertension and renal pathology. However, the mechanisms underlying this increased risk remain unclear. This study utilized an L-NAME-induced preeclampsia mouse model (PELS model) to investigate the effects of PE on offspring blood pressure and renal pathology, focusing on the expression of Angiotensin II Type 1 Receptors (AT1R) and related molecules in renal tissues. Our findings show that L-NAME-induced pre-eclampsia led to reduced birth weights and significantly elevated systolic blood pressure in 6-week-old offspring. Histopathological analysis revealed pronounced glomerular and tubular damage in the kidneys of both 1-week and 6-week-old offspring from the pre-eclampsia group. At 1 week of age, the pre-eclampsia group exhibited elevated mRNA and protein expression levels of AT1R, GRK4, AQP2, ENaC, and NCC in renal tissues compared to controls. However, these differences were no longer significant at 6 weeks of age. No significant gender differences were observed in either blood pressure or renal pathological changes. Preeclampsia induced by L-NAME results in increased blood pressure and renal damage in offspring, potentially mediated by early alterations in the renal RAS system. The observed changes in AT1R and related molecules appear to be transient, suggesting that the early impact of pre-eclampsia on renal structure may trigger, but not sustain, hypertension in offspring. Further studies are needed to elucidate the long-term mechanisms driving hypertension in this population.
Collapse
Affiliation(s)
- Yong Wang
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hao Wang
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Huiqing Lu
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ji Ma
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Wei Wu
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yinan Wang
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bo Ma
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hao Zhu
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| | - Rong Hu
- The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Falkner B, Alexander BT, Nuyt AM, South AM, Ingelfinger J. Cardiovascular Health Starts in the Womb. Hypertension 2024; 81:2016-2026. [PMID: 39069922 PMCID: PMC11410535 DOI: 10.1161/hypertensionaha.124.21359] [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] [Indexed: 07/30/2024]
Abstract
Hypertension has largely been viewed as a disorder of adulthood. Historically, blood pressure (BP) was not routinely measured in children because hypertension was considered uncommon in childhood. It was not until the 1970s that it was apparent that in childhood BP levels were normally lower compared with those in adults, were related to age and growth, and that abnormal BP in children needed different definitions. Based on the distribution of BP levels in available child cohorts, the 95th percentile of BP levels became the definition of hypertension in children and adolescents-an epidemiological definition. Subsequent clinical and epidemiological research identified associated risk factors in childhood that linked abnormal BP in youth with hypertension in adulthood. In the 1980s, the Barker hypothesis, based on observations that low birth weight could be linked to cardiovascular disease in adulthood, promoted further research spanning epidemiological, clinical, and basic science on the childhood origins of hypertension. This review focuses on recent findings from both longitudinal maternal-child cohorts and experimental models that examine both maternal and offspring conditions associated with risks of subsequent cardiovascular disease.
Collapse
Affiliation(s)
- Bonita Falkner
- Departments of Medicine (B.F.), Sydney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA
- Pediatrics (B.F.), Sydney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson (B.T.A.)
| | - Anne-Monique Nuyt
- Department of Pediatrics, CHU Sainte Justine, Faculté de Médecine, Université de Montréal, QC (A.-M.N.)
| | - Andrew M South
- Department of Pediatrics, Section of Nephrology, Wake Forest University School of Medicine, Winston Salem, NC (A.M.S.)
| | - Julie Ingelfinger
- Pediatric Nephrology Unit, MassGeneral Hospital for Children at MassGeneral, Boston, MA (J.I.)
| |
Collapse
|
3
|
Calcaterra V, Mannarino S, Garella V, Rossi V, Biganzoli EM, Zuccotti G. Cardiovascular Risk in Pediatrics: A Dynamic Process during the First 1000 Days of Life. Pediatr Rep 2023; 15:636-659. [PMID: 37987283 PMCID: PMC10661305 DOI: 10.3390/pediatric15040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/08/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023] Open
Abstract
The early childhood period, encompassing prenatal and early stages, assumes a pivotal role in shaping cardiovascular risk factors. We conducted a narrative review, presenting a non-systematic summation and analysis of the available literature, focusing on cardiovascular risk from prenatal development to the first 1000 days of life. Elements such as maternal health, genetic predisposition, inadequate fetal nutrition, and rapid postnatal growth contribute to this risk. Specifically, maternal obesity and antibiotic use during pregnancy can influence transgenerational risk factors. Conditions at birth, such as fetal growth restriction and low birth weight, set the stage for potential cardiovascular challenges. To consider cardiovascular risk in early childhood as a dynamic process is useful when adopting a personalized prevention for future healthcare and providing recommendations for management throughout their journey from infancy to early adulthood. A comprehensive approach is paramount in addressing early childhood cardiovascular risks. By targeting critical periods and implementing preventive strategies, healthcare professionals and policymakers can pave the way for improved cardiovascular outcomes. Investing in children's health during their early years holds the key to alleviating the burden of cardiovascular diseases for future generations.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Savina Mannarino
- Pediatric Cardiology Unit, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Vittoria Garella
- Pediatric Cardiology Unit, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Virginia Rossi
- Pediatric Department, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Elia Mario Biganzoli
- Medical Statistics Unit, Department of Biomedical and Clinical Sciences, University Hospital, University of Milan, 20157 Milan, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children's Hospital, 20154 Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, 20157 Milan, Italy
| |
Collapse
|
4
|
Alexander BT, South AM, August P, Bertagnolli M, Ferranti EP, Grobe JL, Jones EJ, Loria AS, Safdar B, Sequeira-Lopez MLS. Appraising the Preclinical Evidence of the Role of the Renin-Angiotensin-Aldosterone System in Antenatal Programming of Maternal and Offspring Cardiovascular Health Across the Life Course: Moving the Field Forward: A Scientific Statement From the American Heart Association. Hypertension 2023; 80:e75-e89. [PMID: 36951054 PMCID: PMC10242542 DOI: 10.1161/hyp.0000000000000227] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
There is increasing interest in the long-term cardiovascular health of women with complicated pregnancies and their affected offspring. Emerging antenatal risk factors such as preeclampsia appear to increase the risk of hypertension and cardiovascular disease across the life course in both the offspring and women after pregnancy. However, the antenatal programming mechanisms responsible are complex and incompletely understood, with roots in alterations in the development, structure, and function of the kidney, heart, vasculature, and brain. The renin-angiotensin-aldosterone system is a major regulator of maternal-fetal health through the placental interface, as well as kidney and cardiovascular tissue development and function. Renin-angiotensin-aldosterone system dysregulation plays a critical role in the development of pregnancy complications such as preeclampsia and programming of long-term adverse cardiovascular health in both the mother and the offspring. An improved understanding of antenatal renin-angiotensin-aldosterone system programming is crucial to identify at-risk individuals and to facilitate development of novel therapies to prevent and treat disease across the life course. Given the inherent complexities of the renin-angiotensin-aldosterone system, it is imperative that preclinical and translational research studies adhere to best practices to accurately and rigorously measure components of the renin-angiotensin-aldosterone system. This comprehensive synthesis of preclinical and translational scientific evidence of the mechanistic role of the renin-angiotensin-aldosterone system in antenatal programming of hypertension and cardiovascular disease will help (1) to ensure that future research uses best research practices, (2) to identify pressing needs, and (3) to guide future investigations to maximize potential outcomes. This will facilitate more rapid and efficient translation to clinical care and improve health outcomes.
Collapse
|
5
|
Amruta N, Kandikattu HK, Intapad S. Cardiovascular Dysfunction in Intrauterine Growth Restriction. Curr Hypertens Rep 2022; 24:693-708. [PMID: 36322299 DOI: 10.1007/s11906-022-01228-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW We highlight important new findings on cardiovascular dysfunction in intrauterine growth restriction. RECENT FINDINGS Intrauterine growth restriction (IUGR) is a multifactorial condition which negatively impacts neonatal growth during pregnancy and is associated with health problems during the lifespan. It affects 5-15% of all pregnancies in the USA and Europe with varying percentages in developing countries. Epidemiological studies have reported that IUGR is associated with the pathogenesis of hypertension, activation of the renin-angiotensin system (RAS), disruption in placental-mTORC and TGFβ signaling cascades, and endothelial dysfunction in IUGR fetuses, children, adolescents, and adults resulting in the development of cardiovascular diseases (CVD). Experimental studies are needed to investigate therapeutic measures to treat increased blood pressure (BP) and long-term CVD problems in people affected by IUGR. We outline the mechanisms mediating fetal programming of hypertension in developing CVD. We have reviewed findings from different experimental models focusing on recent studies that demonstrate CVD in IUGR.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, #8683, New Orleans, LA, 70112-2699, USA
| | - Hemanth Kumar Kandikattu
- Department of Medicine, Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Suttira Intapad
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, #8683, New Orleans, LA, 70112-2699, USA.
| |
Collapse
|
6
|
Ashraf UM, Hall DL, Campbell N, Waller JP, Rawls AZ, Solise D, Cockrell K, Bidwell GL, Romero DG, Ojeda NB, LaMarca B, Alexander BT. Inhibition of the AT 1R agonistic autoantibody in a rat model of preeclampsia improves fetal growth in late gestation. Am J Physiol Regul Integr Comp Physiol 2022; 323:R670-R681. [PMID: 36121142 PMCID: PMC9602704 DOI: 10.1152/ajpregu.00122.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/17/2022] [Accepted: 09/09/2022] [Indexed: 02/07/2023]
Abstract
Placenta ischemia, the initiating event in preeclampsia (PE), is associated with fetal growth restriction. Inhibition of the agonistic autoantibody against the angiotensin type 1 receptor AT1-AA, using an epitope-binding inhibitory peptide ('n7AAc') attenuates increased blood pressure at gestational day (G)19 in the clinically relevant reduced uterine perfusion pressure (RUPP) model of PE. Thus we tested the hypothesis that maternal administration of 'n7AAc' does not transfer to the fetus, improves uterine blood flow and fetal growth, and attenuates elevated placental expression of miRNAs implicated in PE and FGR. Sham or RUPP surgery was performed at G14 with vehicle or 'n7AAc' (144 µg/day) administered via an osmotic pump from G14 to G20. Maternal plasma levels of the peptide on G20 were 16.28 ± 4.4 nM, and fetal plasma levels were significantly lower at 1.15 ± 1.7 nM (P = 0.0007). The uterine artery resistance index was significantly elevated in RUPP (P < 0.0001) but was not increased in 'n7AAc'-RUPP or 'n7AAc'-Sham versus Sham. A significant reduction in fetal weight at G20 in RUPP (P = 0.003) was not observed in 'n7AAc'-RUPP. Yet, percent survival was reduced in RUPP (P = 0.0007) and 'n7AAc'-RUPP (P < 0.0002). Correlation analysis indicated the reduction in percent survival during gestation was specific to the RUPP (r = 0.5342, P = 0.043) and independent of 'n7AAc'. Placental miR-155 (P = 0.0091) and miR-181a (P = 0.0384) expression was upregulated in RUPP at G20 but was not elevated in 'n7AAc'-RUPP. Collectively, our results suggest that maternal administration of 'n7AAc' does not alter fetal growth in the RUPP implicating its potential as a therapeutic for the treatment of PE.NEW & NOTEWORTHY The seven amino acid inhibitory peptide to the AT1-AA ('n7AAc') has limited transfer to the fetus at gestational day 20, improves uterine blood flow and fetal growth in the reduced uterine perfusion pressure model of preeclampsia (PE), and does not impair fetal survival during gestation in sham-operated or placental ischemic rats. Collectively, these findings suggest that maternal administration of 'n7AAc' as an effective strategy for the treatment of PE is associated with improved outcomes in the fetus.
Collapse
Affiliation(s)
- Usman M Ashraf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jamarius P Waller
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Adam Z Rawls
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dylan Solise
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kathy Cockrell
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
7
|
Tain YL, Hsu CN. Developmental and Early Life Origins of Hypertension: Preventive Aspects of Melatonin. Antioxidants (Basel) 2022; 11:924. [PMID: 35624788 PMCID: PMC9138087 DOI: 10.3390/antiox11050924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Hypertension represents a major disease burden worldwide. Abundant evidence suggests that hypertension can originate in early life. Adverse programming processes can be prevented by early life intervention-namely, reprogramming-to avoid developing chronic diseases later in life. Melatonin is an endogenously produced hormone with a multifaceted biological function. Although melatonin supplementation has shown benefits for human health, less attention has been paid to exploring its reprogramming effects on the early life origins of hypertension. In this review, first, we discuss the physiological roles of melatonin in pregnancy, fetal development, and the regulation of blood pressure. Then, we summarize the epidemiological and experimental evidence for the early life origins of hypertension. This is followed by a description of the animal models used to examine early melatonin therapy as a reprogramming strategy to protect against the early life origins of hypertension. A deeper understanding of the developmental programming of hypertension and recent advances in early melatonin intervention might provide a path forward in reducing the global burden of hypertension.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Hsu CN, Tain YL. Chronic Kidney Disease and Gut Microbiota: What Is Their Connection in Early Life? Int J Mol Sci 2022; 23:3954. [PMID: 35409313 PMCID: PMC9000069 DOI: 10.3390/ijms23073954] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The gut-kidney interaction implicating chronic kidney disease (CKD) has been the focus of increasing interest in recent years. Gut microbiota-targeted therapies could prevent CKD and its comorbidities. Considering that CKD can originate in early life, its treatment and prevention should start in childhood or even earlier in fetal life. Therefore, a better understanding of how the early-life gut microbiome impacts CKD in later life and how to develop ideal early interventions are unmet needs to reduce CKD. The purpose of the current review is to summarize (1) the current evidence on the gut microbiota dysbiosis implicated in pediatric CKD; (2) current knowledge supporting the impact of the gut-kidney axis in CKD, including inflammation, immune response, alterations of microbiota compositions, short-chain fatty acids, and uremic toxins; and (3) an overview of the studies documenting early gut microbiota-targeted interventions in animal models of CKD of developmental origins. Treatment options include prebiotics, probiotics, postbiotics, etc. To accelerate the transition of gut microbiota-based therapies for early prevention of CKD, an extended comprehension of gut microbiota dysbiosis implicated in renal programming is needed, as well as a greater focus on pediatric CKD for further clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
9
|
Awazu M. Structural and functional changes in the kidney caused by adverse fetal and neonatal environments. Mol Biol Rep 2021; 49:2335-2344. [PMID: 34817775 DOI: 10.1007/s11033-021-06967-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
Health and disease risk in the adulthood are known to be affected by the early developmental environment. Kidney diseases are one of these diseases, and kidneys are altered both structurally and functionally by adverse pre- and perinatal events. The most known structural change is low nephron number seen in subjects born low birth weight and/or preterm. In various animal models of intrauterine growth restriction (IUGR), one of the causes of low birth weight, the mechanism of low nephron number was investigated. While apoptosis of metanephric mesenchyme has been suggested to be the cause, I showed that suppression of ureteric branching, global DNA methylation, and caspase-3 activity also contributes to the mechanism. Other structural changes caused by adverse fetal and neonatal environments include peritubular and glomerular capillary rarefaction and low podocyte endowment. These are aggravated by postnatal development of focal glomerulosclerosis and tubulointerstitial fibrosis that result from low nephron number. Functional changes can be seen in tubules, endothelium, renin-angiotensin system, sympathetic nervous system, oxidative stress, and others. As an example, I reported that aggravated nitrosative stress in a rat IUGR model resulted in more severe tubular necrosis and tubulointerstitial fibrosis after unilateral ureteral obstruction. The mechanism of various functional changes needs to be clarified but may be explained by epigenetic modifications.
Collapse
Affiliation(s)
- Midori Awazu
- Department of Pediatrics, Tokyo Metropolitan Ohtsuka Hospital, Tokyo, Japan.
| |
Collapse
|
10
|
Hsu CN, Hou CY, Hsu WH, Tain YL. Early-Life Origins of Metabolic Syndrome: Mechanisms and Preventive Aspects. Int J Mol Sci 2021; 22:11872. [PMID: 34769303 PMCID: PMC8584419 DOI: 10.3390/ijms222111872] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
One of the leading global public-health burdens is metabolic syndrome (MetS), despite the many advances in pharmacotherapies. MetS, now known as "developmental origins of health and disease" (DOHaD), can have its origins in early life. Offspring MetS can be programmed by various adverse early-life conditions, such as nutrition imbalance, maternal conditions or diseases, maternal chemical exposure, and medication use. Conversely, early interventions have shown potential to revoke programming processes to prevent MetS of developmental origins, namely reprogramming. In this review, we summarize what is currently known about adverse environmental insults implicated in MetS of developmental origins, including the fundamental underlying mechanisms. We also describe animal models that have been developed to study the developmental programming of MetS. This review extends previous research reviews by addressing implementation of reprogramming strategies to prevent the programming of MetS. These mechanism-targeted strategies include antioxidants, melatonin, resveratrol, probiotics/prebiotics, and amino acids. Much work remains to be accomplished to determine the insults that could induce MetS, to identify the mechanisms behind MetS programming, and to develop potential reprogramming strategies for clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan;
| | - Wei-Hsuan Hsu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Chen Kung University, Tainan 701, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
11
|
Bhunu B, Riccio I, Intapad S. Insights into the Mechanisms of Fetal Growth Restriction-Induced Programming of Hypertension. Integr Blood Press Control 2021; 14:141-152. [PMID: 34675650 PMCID: PMC8517636 DOI: 10.2147/ibpc.s312868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/30/2021] [Indexed: 12/21/2022] Open
Abstract
In recent decades, both clinical and animal studies have shown that fetal growth restriction (FGR), caused by exposure to adverse uterine environments, is a risk factor for hypertension as well as for a variety of adult diseases. This observation has shaped and informed the now widely accepted theory of developmental origins of health and disease (DOHaD). There is a plethora of evidence supporting the association of FGR with increased risk of adult hypertension; however, the underlying mechanisms responsible for this correlation remain unclear. This review aims to explain the current advances in the field of fetal programming of hypertension and a brief narration of the underlying mechanisms that may link FGR to increased risk of adult hypertension. We explain the theory of DOHaD and then provide evidence from both clinical and basic science research which support the theory of fetal programming of adult hypertension. In addition, we have explored the underlying mechanisms that may link FGR to an increased risk of adult hypertension. These mechanisms include epigenetic changes, metabolic disorders, vascular dysfunction, neurohormonal impairment, and alterations in renal physiology and function. We further describe sex differences seen in the developmental origins of hypertension and provide insights into the opportunities and challenges present in this field.
Collapse
Affiliation(s)
- Benjamin Bhunu
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Isabel Riccio
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Suttira Intapad
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| |
Collapse
|
12
|
Wojczakowski W, Kimber-Trojnar Ż, Dziwisz F, Słodzińska M, Słodziński H, Leszczyńska-Gorzelak B. Preeclampsia and Cardiovascular Risk for Offspring. J Clin Med 2021; 10:jcm10143154. [PMID: 34300320 PMCID: PMC8306208 DOI: 10.3390/jcm10143154] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
There is growing evidence of long-term cardiovascular sequelae in children after in utero exposure to preeclampsia. Maternal hypertension and/or placental ischaemia during pregnancy increase the risk of hypertension, stroke, diabetes, and cardiovascular disease (CVD) in the offspring later in life. The mechanisms associated with CVD seem to be a combination of genetic, molecular, and environmental factors which can be defined as fetal and postnatal programming. The aim of this paper is to discuss the relationship between pregnancy complicated by preeclampsia and possibility of CVD in the offspring. Unfortunately, due to its multifactorial nature, a clear dependency mechanism between preeclampsia and CVD is difficult to establish.
Collapse
Affiliation(s)
- Wiktor Wojczakowski
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| | - Żaneta Kimber-Trojnar
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
- Correspondence: ; Tel.: +48-81-7244-769
| | - Filip Dziwisz
- Department of Interventional Cardiology and Cardiac Arrhythmias, Medical University of Lodz, 90-549 Łódź, Poland;
| | - Magdalena Słodzińska
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| | - Hubert Słodziński
- Institute of Medical Sciences, State School of Higher Education in Chełm, 22-100 Chełm, Poland;
| | - Bożena Leszczyńska-Gorzelak
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| |
Collapse
|
13
|
de Souza AMA, Ecelbarger CM, Sandberg K. Caloric Restriction and Cardiovascular Health: the Good, the Bad, and the Renin-Angiotensin System. Physiology (Bethesda) 2021; 36:220-234. [PMID: 34159807 DOI: 10.1152/physiol.00002.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Much excitement exists over the cardioprotective and life-extending effects of caloric restriction (CR). This review integrates population studies with experimental animal research to address the positive and negative impact of mild and severe CR on cardiovascular physiology and pathophysiology, with a particular focus on the renin-angiotensin system (RAS). We also highlight the gaps in knowledge and areas ripe for future physiological research.
Collapse
Affiliation(s)
- Aline M A de Souza
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Carolyn M Ecelbarger
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Kathryn Sandberg
- Division of Nephrology and Hypertension, Department of Medicine, Georgetown University, Washington, District of Columbia
| |
Collapse
|
14
|
Coats LE, Bakrania BA, Bamrick-Fernandez DR, Ariatti AM, Rawls AZ, Ojeda NB, Alexander BT. Soluble guanylate cyclase stimulation in late gestation does not mitigate asymmetric intrauterine growth restriction or cardiovascular risk induced by placental ischemia in the rat. Am J Physiol Heart Circ Physiol 2021; 320:H1923-H1934. [PMID: 33739156 DOI: 10.1152/ajpheart.00033.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stimulation of soluble guanylate cyclase (sGC) improves fetal growth at gestational day 20 in the reduced uterine perfusion pressure (RUPP) rat model of placental ischemia suggesting a role for sGC in the etiology of intrauterine growth restriction (IUGR). This study tested the hypothesis that stimulation of sGC until birth attenuates asymmetric IUGR mitigating increased cardiovascular risk in offspring. Sham or RUPP surgery was performed at gestational day 14 (G14); vehicle or the sGC stimulator Riociguat (10 mg/kg/day sc) was administered G14 until birth. Birth weight was reduced in offspring from RUPP [intrauterine growth restricted (IUGR)], sGC RUPP (sGC IUGR), and sGC Sham (sGC Control) compared with Sham (Control). Crown circumference was maintained, but abdominal circumference was reduced in IUGR and sGC IUGR compared with Control indicative of asymmetrical growth. Gestational length was prolonged in sGC RUPP, and survival at birth was reduced in sGC IUGR. Probability of survival to postnatal day 2 was also significantly reduced in IUGR and sGC IUGR versus Control and in sGC IUGR versus IUGR. At 4 mo of age, blood pressure was increased in male IUGR and sGC IUGR but not male sGC Control born with symmetrical IUGR. Global longitudinal strain was increased and stroke volume was decreased in male IUGR and sGC IUGR compared with Control. Thus late gestational stimulation of sGC does not mitigate asymmetric IUGR or increased cardiovascular risk in male sGC IUGR. Furthermore, late gestational stimulation of sGC is associated with symmetrical growth restriction in sGC Control implicating contraindications in normal pregnancy.NEW & NOTEWORTHY The importance of the soluble guanylate cyclase-cGMP pathway in a rat model of placental ischemia differs during critical windows of development, implicating other factors may be critical mediators of impaired fetal growth in the final stages of gestation. Moreover, increased blood pressure at 4 mo of age in male intrauterine growth restriction offspring is associated with impaired cardiac function including an increase in global longitudinal strain in conjunction with a decrease in stroke volume, ejection fraction, and cardiac output.
Collapse
Affiliation(s)
- Laura E Coats
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bhavisha A Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,The University of Queensland Centre for Clinical Research and Perinatal Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Allison M Ariatti
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Adam Z Rawls
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara T Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
15
|
Current Updates on Pre-eclampsia: Maternal and Foetal Cardiovascular Diseases Predilection, Science or Myth? : Future cardiovascular disease risks in mother and child following pre-eclampsia. Curr Hypertens Rep 2021; 23:16. [PMID: 33694011 DOI: 10.1007/s11906-021-01132-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Cardiovascular diseases (CVD), including pre-eclampsia (PE), remain the major cause of death and morbidity in women. This review elucidates the current knowledge, state of research and scientific information available on the post-event implications and complications of PE regarding maternal and foetal cardiovascular health. Does PE expose, predispose or aggravate a predilection to maternal and foetal CVD later in life? RECENT FINDINGS Women with a history of PE are reported to have stiffer arteries and are more likely to develop cardiovascular problems with time, especially aortic stenosis and mitral regurgitation, which were not hitherto linked with hypertensive pregnancy. Foetal cells persistence in the mother long after pregnancy, now clearly established in the lungs of mice postpartum, is suggested to portend an overexpression of STOX1, which may potentiate later life CVD. Moreover, the conventional theories of in utero stress and developmental reprogramming may not adequately explain the risk of later life CVD predilection in offspring born to mothers with pre-eclampsia as recent data has shown that siblings of offspring born from pre-eclamptic pregnancies are also at higher risk of hypertension later in life, irrespective of whether subsequent pregnancies were pre-eclamptic or normotensive. The mechanism involved in adverse cardiovascular outcome in offspring of pre-eclamptic pregnancies is most likely an intricate interaction of foetal programming, environmental and genetic factors. In light of available evidence, the question of whether PE is just a pointer or predisposing factor to maternal development of CVDs in later life begs for answers to facilitate definitive clinical solutions and preventive approaches.
Collapse
|
16
|
Hsu CN, Tain YL. Targeting the Renin-Angiotensin-Aldosterone System to Prevent Hypertension and Kidney Disease of Developmental Origins. Int J Mol Sci 2021; 22:ijms22052298. [PMID: 33669059 PMCID: PMC7956566 DOI: 10.3390/ijms22052298] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is implicated in hypertension and kidney disease. The developing kidney can be programmed by various early-life insults by so-called renal programming, resulting in hypertension and kidney disease in adulthood. This theory is known as developmental origins of health and disease (DOHaD). Conversely, early RAAS-based interventions could reverse program processes to prevent a disease from occurring by so-called reprogramming. In the current review, we mainly summarize (1) the current knowledge on the RAAS implicated in renal programming; (2) current evidence supporting the connections between the aberrant RAAS and other mechanisms behind renal programming, such as oxidative stress, nitric oxide deficiency, epigenetic regulation, and gut microbiota dysbiosis; and (3) an overview of how RAAS-based reprogramming interventions may prevent hypertension and kidney disease of developmental origins. To accelerate the transition of RAAS-based interventions for prevention of hypertension and kidney disease, an extended comprehension of the RAAS implicated in renal programming is needed, as well as a greater focus on further clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-975-056-995; Fax: +886-7733-8009
| |
Collapse
|
17
|
Chen L, Yue J, Zhou S, Hu Y, Li J. Ouabain Protects Nephrogenesis in Rats Experiencing Intrauterine Growth Restriction and Partially Restores Renal Function in Adulthood. Reprod Sci 2021; 28:186-196. [PMID: 32767217 DOI: 10.1007/s43032-020-00280-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022]
Abstract
Intrauterine growth restriction (IUGR) is, in general, accompanied by a reduction of the nephron number, which increases the risk of hypertension and renal dysfunction. Studies have revealed that ouabain can partially restore the number of nephrons during IUGR. However, there is limited information regarding the melioration of nephric structure and function. We used maternal malnutrition to induce an IUGR model in rats. Subsequently, we used a mini-pump to administer ouabain to IUGR rats during pregnancy. Male offspring were divided randomly into two groups. One group was fed a normal diet, whereas the other was fed an isocaloric 8% high-salt diet. Maternal malnutrition led to a reduction in the birth weight and number of nephrons in offspring. At the end of a 40-week follow-up period, offspring from the IUGR group had high blood pressure and abnormal excretion of urinary protein; these parameters were exacerbated in offspring fed a high-salt diet. However, ouabain administration during pregnancy could partially restore the number of nephrons in IUGR offspring, normalize blood pressure, and reduce urinary protein excretion, even when challenged with a high-salt diet. Pathology findings revealed that IUGR, particularly following feeding of a high-salt diet, damaged the ultrastructure of glomeruli, but these harmful effects were ameliorated in offspring treated with ouabain. Collectively, our data suggest that ouabain could rescue nephrogenesis in IUGR newborns and protect (at least in part) the structure and function of the kidney during adulthood even when encountering unfavorable environmental challenges in subsequent life.
Collapse
Affiliation(s)
- Liang Chen
- Department of Gynecology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), 300 Guangzhou Road, Nanjing, 210000, China.
| | - Jing Yue
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital (The Affiliated Hospital of Nanjing University Medical School), Nanjing, China
| | - Shulin Zhou
- Department of Gynecology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), 300 Guangzhou Road, Nanjing, 210000, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital (The Affiliated Hospital of Nanjing University Medical School), Nanjing, China
| | - Juan Li
- Department of Hematology, Nanjing Drum Tower Hospital (The Affiliated Hospital of Nanjing University Medical School), Nanjing, China
| |
Collapse
|
18
|
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: 35] [Impact Index Per Article: 7.0] [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
|
19
|
Fetal Growth Restriction and Hypertension in the Offspring: Mechanistic Links and Therapeutic Directions. J Pediatr 2020; 224:115-123.e2. [PMID: 32450071 PMCID: PMC8086836 DOI: 10.1016/j.jpeds.2020.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
20
|
McArdle Z, Schreuder MF, Moritz KM, Denton KM, Singh RR. Physiology and Pathophysiology of Compensatory Adaptations of a Solitary Functioning Kidney. Front Physiol 2020; 11:725. [PMID: 32670095 PMCID: PMC7332829 DOI: 10.3389/fphys.2020.00725] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Children born with a solitary functioning kidney (SFK) have an increased risk of hypertension and kidney disease from early in adulthood. In response to a reduction in kidney mass, the remaining kidney undergoes compensatory kidney growth. This is associated with both an increase in size of the kidney tubules and the glomeruli and an increase in single nephron glomerular filtration rate (SNGFR). The compensatory hypertrophy and increase in filtration at the level of the individual nephron results in normalization of total glomerular filtration rate (GFR). However, over time these same compensatory mechanisms may contribute to kidney injury and hypertension. Indeed, approximately 50% of children born with a SFK develop hypertension by the age of 18 and 20–40% require dialysis by the age of 30. The mechanisms that result in kidney injury are only partly understood, and early biomarkers that distinguish those at an elevated risk of kidney injury are needed. This review will outline the compensatory adaptations to a SFK, and outline how these adaptations may contribute to kidney injury and hypertension later in life. These will be based largely on the mechanisms we have identified from our studies in an ovine model of SFK, that implicate the renal nitric oxide system, the renin angiotensin system and the renal nerves to kidney disease and hypertension associated with SFK. This discussion will also evaluate current, and speculate on next generation, prognostic factors that may predict those children at a higher risk of future kidney disease and hypertension.
Collapse
Affiliation(s)
- Zoe McArdle
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Kate M Denton
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Reetu R Singh
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Insights into sympathetic nervous system and GPCR interplay in fetal programming of hypertension: a bridge for new pharmacological strategies. Drug Discov Today 2020; 25:739-747. [PMID: 32032706 DOI: 10.1016/j.drudis.2020.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death from noncommunicable diseases worldwide. In addition to the classical CVD risk factors related to lifestyle and/or genetic background, exposure to an adverse intrauterine environment compromises fetal development leading to low birth weight and increasing offspring susceptibility to develop CVDs later in life, particularly hypertension - a process known as fetal programming of hypertension (FPH). In FPH animal models, permanent alterations have been detected in gene expression, in the structure and function of heart and blood vessels, compromising cardiovascular physiology and favoring hypertension development. This review focuses on the role of the sympathetic nervous system and its interplay with G-protein-coupled receptors, emphasizing strategies that envisage the prevention and/or treatment of FPH through interventions in early life.
Collapse
|
22
|
Kooiman J, Terstappen F, van Wagensveld L, Franx A, Wever KE, Roseboom TJ, Joles JA, Gremmels H, Lely AT. Conflicting Effects of Fetal Growth Restriction on Blood Pressure Between Human and Rat Offspring: A Meta-Analysis. Hypertension 2020; 75:806-818. [PMID: 31983304 DOI: 10.1161/hypertensionaha.119.14111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Low birth weight is associated with hypertension. Low birth weight can result from fetal growth restriction (FGR) or prematurity. FGR is postulated to impact blood pressure (BP) by developmental programming. This systematic review and meta-analysis studies BP in human and animal offspring following FGR. Pubmed and Web of Science were searched for studies reporting on BP after placental insufficiency induced FGR compared with normal growth controls. Primary outcome was mean absolute BP difference (ΔBP mm Hg [95% CI]). Meta-analysis was performed using random-effects models. Subgroup analyses were executed on species, sex, age, pregnancy duration, and stress during BP readings. Due to large interspecies heterogeneity, analyses were performed separately for human (n=41) and animal (n=31) studies, the latter restricted to rats (n=27). Human studies showed a ΔBP between FGR and controls of -0.6 mm Hg ([95% CI, -1.7 to 0.6]; I2=91%). Mean ΔBP was -2.6 mm Hg (95% CI, -5.7 to 0.4) in women versus -0.5 mm Hg (95% CI, -3.7 to 2.7) in men. Subgroup analyses did not indicate age, gestational age, and stress during measurements as sources of heterogeneity. In rats, mean BP was 12.0 mm Hg ([95% CI, 8.8-15.2]; I2=81%) higher in FGR offspring. This difference was more pronounced in FGR males (13.6 mm Hg [95% CI, 10.3-17.0] versus 9.1 mm Hg [95% CI, 5.3-12.8]). Subgroup analyses on age showed no statistical interaction. BP readings under restrained conditions resulted in larger BP differences between FGR and control rats (15.3 mm Hg [95% CI, 11.6-18.9] versus 5.7 mm Hg [95% CI, 1.1-10.3]). Rat studies confirm the relation between FGR and offspring BP, while observational studies in humans do not show such differences. This may be due to the observational nature of human studies, methodological limitations, or an absence of this phenomenon in humans. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: CRD42018091819.
Collapse
Affiliation(s)
- Judith Kooiman
- From the Department of Obstetrics (J.K., F.T., L.v.W., A.F., A.T.L.), University Medical Center Utrecht, the Netherlands
| | - Fieke Terstappen
- From the Department of Obstetrics (J.K., F.T., L.v.W., A.F., A.T.L.), University Medical Center Utrecht, the Netherlands.,Department of Developmental Origin of Disease (F.T.), University Medical Center Utrecht, the Netherlands
| | - Lilian van Wagensveld
- From the Department of Obstetrics (J.K., F.T., L.v.W., A.F., A.T.L.), University Medical Center Utrecht, the Netherlands
| | - Arie Franx
- From the Department of Obstetrics (J.K., F.T., L.v.W., A.F., A.T.L.), University Medical Center Utrecht, the Netherlands
| | - Kimberley E Wever
- Systematic Review Center for Laboratory animal Experimentation (SYRCLE), Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands (K.E.W.)
| | - Tessa J Roseboom
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, the Netherlands (T.J.R.)
| | - Jaap A Joles
- Wilhelmina Children's Hospital and Department of Nephrology and Hypertension (J.A.J., H.G.), University Medical Center Utrecht, the Netherlands
| | - Hendrik Gremmels
- Wilhelmina Children's Hospital and Department of Nephrology and Hypertension (J.A.J., H.G.), University Medical Center Utrecht, the Netherlands
| | - A Titia Lely
- From the Department of Obstetrics (J.K., F.T., L.v.W., A.F., A.T.L.), University Medical Center Utrecht, the Netherlands
| |
Collapse
|
23
|
Association between preterm birth and the renin-angiotensin system in adolescence: influence of sex and obesity. J Hypertens 2019; 36:2092-2101. [PMID: 29846325 DOI: 10.1097/hjh.0000000000001801] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Preterm birth appears to contribute to early development of cardiovascular disease, but the mechanisms are unknown. Prematurity may result in programming events that alter the renin-angiotensin system. We hypothesized that prematurity is associated with lower angiotensin-(1-7) in adolescence and that sex and obesity modify this relationship. METHODS We quantified angiotensin II and angiotensin-(1-7) in the plasma and urine of 175 adolescents born preterm and 51 term-born controls. We used generalized linear models to estimate the association between prematurity and the peptides, controlling for confounding factors and stratifying by sex and overweight/obesity. RESULTS Prematurity was associated with lower plasma angiotensin II (β: -5.2 pmol/l, 95% CI: -10.3 to -0.04) and angiotensin-(1-7) (-5.2 pmol/l, 95% CI: -8.4 to -2.0) but overall higher angiotensin II:angiotensin-(1-7) (3.0, 95% CI: 0.9-5.0). The preterm-term difference in plasma angiotensin-(1-7) was greater in women (-6.9 pmol/l, 95% CI: -10.7 to -3.1) and individuals with overweight/obesity (-8.0 pmol/l, 95% CI: -12.2 to -3.8). The preterm-term difference in angiotensin II:angiotensin-(1-7) was greater among those with overweight/obesity (4.4, 95% CI: 0.6-8.1). On multivariate analysis, prematurity was associated with lower urinary angiotensin II:angiotensin-(1-7) (-0.13, 95% CI: -0.26 to -0.003), especially among the overweight/obesity group (-0.38, 95% CI: -0.72 to -0.04). CONCLUSION Circulating angiotensin-(1-7) was diminished whereas urinary angiotensin-(1-7) was increased relative to angiotensin II in adolescents born preterm, suggesting prematurity may increase the risk of cardiovascular disease by altering the renin-angiotensin system. Perinatal renin-angiotensin system programming was more pronounced in women and individuals with overweight/obesity, thus potentially augmenting their risk of developing early cardiovascular disease.
Collapse
|
24
|
Lu HQ, Hu R. Lasting Effects of Intrauterine Exposure to Preeclampsia on Offspring and the Underlying Mechanism. AJP Rep 2019; 9:e275-e291. [PMID: 31511798 PMCID: PMC6736667 DOI: 10.1055/s-0039-1695004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a common pregnancy complication which can have adverse impact on both mother and baby. In addition to the short term effects, a large body of epidemiological evidence has found preeclampsia can exert long-lasting effects on mother and offspring. Studies suggest that offspring exposed to preeclampsia are at a higher risk of developing cardiovascular, metabolic, and neurological diseases, as well as other diseases. However, studies investigating the underlying mechanism are limited, the exact mechanism still remains unclear. In this study, we will review the epidemiological evidence and studies exploring the mechanism underlying long-term effects of preeclampsia on offspring. Further studies should be targeted at this field so as to implement effective clinical management to prevent the exposed offspring from potential diseases.
Collapse
Affiliation(s)
- Hui Qing Lu
- Department of Obstetrics, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Rong Hu
- Department of Obstetrics, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Gallo LA, Walton SL, Mazzuca MQ, Tare M, Parkington HC, Wlodek ME, Moritz KM. Uteroplacental insufficiency temporally exacerbates salt-induced hypertension associated with a reduced natriuretic response in male rat offspring. J Physiol 2018; 596:5859-5872. [PMID: 29604087 PMCID: PMC6265551 DOI: 10.1113/jp275655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/21/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Low weight at birth increases the risk of developing chronic diseases in adulthood A diet that is high in salt is known to elevate blood pressure, which is a major risk factor for cardiovascular and kidney diseases The present study demonstrates that growth restricted male rats have a heightened sensitivity to high dietary salt, in the context of raised systolic blood pressure, reduced urinary sodium excretion and stiffer mesenteric resistance vessels Other salt-induced effects, such as kidney hyperfiltration, albuminuria and glomerular damage, were not exacerbated by being born small The present study demonstrates that male offspring born small have an increased cardiovascular susceptibility to high dietary salt, such that that minimizing salt intake is probably of particular benefit to this at-risk population ABSTRACT: Intrauterine growth restriction increases the risk of developing chronic diseases in adulthood. Lifestyle factors, such as poor dietary choices, may elevate this risk. We determined whether being born small increases the sensitivity to a dietary salt challenge, in the context of hypertension, kidney disease and arterial stiffness. Bilateral uterine vessel ligation or sham surgery (offspring termed Restricted and Control, respectively) was performed on 18-day pregnant Wistar Kyoto rats. Male offspring were allocated to receive a diet high in salt (8% sodium chloride) or remain on standard rat chow (0.52% sodium chloride) from 20 to 26 weeks of age for 6 weeks. Systolic blood pressure (tail-cuff), renal function (24 h urine excretions) and vascular stiffness (pressure myography) were assessed. Restricted males were born 15% lighter than Controls and remained smaller throughout the study. Salt-induced hypertension was exacerbated in Restricted offspring, reaching a peak systolic pressure of ∼175 mmHg earlier than normal weight counterparts. The natriuretic response to high dietary salt in Restricted animals was less than in Controls and may explain the early rise in arterial pressure. Growth restricted males allocated to a high salt diet also had increased passive arterial stiffness of mesenteric resistance arteries. Other aspects of renal function, including salt-induced hyperfiltration, albuminuria and glomerular damage, were not exacerbated by uteroplacental insufficiency. The present study demonstrates that male offspring exposed to uteroplacental insufficiency and born small have an increased sensitivity to salt-induced hypertension and arterial remodelling.
Collapse
Affiliation(s)
- Linda A. Gallo
- Department of PhysiologyThe University of MelbourneVICAustralia
- School of Biomedical SciencesThe University of QueenslandQLDAustralia
- Mater Research Institute‐The University of QueenslandTranslational Research InstituteQLDAustralia
| | - Sarah L. Walton
- School of Biomedical SciencesThe University of QueenslandQLDAustralia
- Child Health Research CentreThe University of QueenslandQLDAustralia
| | - Marc Q. Mazzuca
- Department of PhysiologyThe University of MelbourneVICAustralia
| | - Marianne Tare
- Department of PhysiologyMonash UniversityVICAustralia
- Monash Rural HealthMonash UniversityVICAustralia
| | | | - Mary E. Wlodek
- Department of PhysiologyThe University of MelbourneVICAustralia
| | - Karen M. Moritz
- School of Biomedical SciencesThe University of QueenslandQLDAustralia
- Child Health Research CentreThe University of QueenslandQLDAustralia
| |
Collapse
|
26
|
Brennan LJ, Goulopoulou S, Bourque SL. Prenatal therapeutics and programming of cardiovascular function. Pharmacol Res 2018; 139:261-272. [PMID: 30458216 DOI: 10.1016/j.phrs.2018.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVD) are a leading cause of mortality worldwide. Despite recognizing the importance of risk factors in dictating CVD susceptibility and onset, patient treatment remains a challenging endeavor. Increasingly, the benefits of prevention and mitigation of risk factors earlier in life are being acknowledged. The developmental origins of health and disease posits that insults during specific periods of development can influence long-term health outcomes; this occurs because the developing organism is highly plastic, and hence vulnerable to environmental perturbations. By extension, targeted therapeutics instituted during critical periods of development may confer long-term protection, and thus reduce the risk of CVD in later life. This review provides a brief overview of models of developmental programming, and then discusses the impact of perinatal therapeutic interventions on long-term cardiovascular function in the offspring. The discussion focuses on bioactive food components, as well as pharmacological agents currently approved for use in pregnancy; in short, those agents most likely to be used in pregnancy and early childhood.
Collapse
Affiliation(s)
- Lesley J Brennan
- Department of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Canada.
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, United States.
| | - Stephane L Bourque
- Department of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Canada.
| |
Collapse
|
27
|
Yeung KR, Sunderland N, Lind JM, Heffernan S, Pears S, Xu B, Hennessy A, Makris A. Increased salt sensitivity in offspring of pregnancies complicated by experimental preeclampsia. Clin Exp Pharmacol Physiol 2018; 45:1302-1308. [PMID: 29992611 DOI: 10.1111/1440-1681.13008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 01/28/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy known to increase the risk of cardiovascular disease in mothers and offspring. Offspring exposed to a suboptimal intrauterine environment may experience altered fetal programming and subsequent long-term cardiovascular changes. This study investigated changes in the vascular response in offspring from experimental preeclampsia (EPE) induced by uterine artery ligation, in the absence of fetal growth restriction, compared to normal baboon pregnancies (controls), following a high salt diet challenge. After 1 week of standard diet (containing <1% salt), animals were fed a high salt diet (6%) for 2 weeks. Systolic and diastolic blood pressure (SBP, DBP), aldosterone, renin and creatinine clearance were evaluated in EPE (n = 6, 50% male) and control (n = 6, 50% male) offspring. A repeated measures analysis was performed, and P < 0.05 was considered significant. At baseline, there were no differences between the groups in any parameter (EPE, mean age and weight 3.2 ± 1.2 years, 6.8 ± 1.0 kg, respectively; Control, 2.9 ± 0.8 years, 7.1 ± 1.5 kg). After salt loading the EPE group had significantly higher SBP (92 ± 5 mm Hg) compared to the control group (83 ± 4 mm Hg, P = 0.03). Aldosterone concentration was higher in the EPE group despite the same salt excretion and no difference in renal function. Salt sensitivity may differ in offspring from hypertensive pregnancies due to fetal programming. This could have long-term consequences for cardiovascular health of EPE offspring and further research is required to determine the exact pathological mechanisms.
Collapse
Affiliation(s)
- Kristen R Yeung
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | | | | | | | - Suzanne Pears
- Heart Research Institute, Sydney, NSW, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Bei Xu
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | - Annemarie Hennessy
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia
| | - Angela Makris
- Western Sydney University, Sydney, NSW, Australia.,Heart Research Institute, Sydney, NSW, Australia.,Nephrology Department, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
28
|
Up-regulation of renal renin-angiotensin system and inflammatory mechanisms in the prenatal programming by low-protein diet: beneficial effect of the post-weaning losartan treatment. J Dev Orig Health Dis 2018; 9:530-535. [PMID: 29729681 DOI: 10.1017/s2040174418000296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Previous studies have shown that the renin-angiotensin system (RAS) is affected by adverse maternal nutrition during pregnancy. The aim of this study was to investigate the effects of a maternal low-protein diet on proinflammatory cytokines, reactive oxygen species and RAS components in kidney samples isolated from adult male offspring. We hypothesized that post-weaning losartan treatment would have beneficial effects on RAS activity and inflammatory and oxidative stress markers in these animals. Pregnant Sprague-Dawley rats were fed with a control (20% casein) or low-protein diet (LP) (6% casein) throughout gestation. After weaning, the LP pups were randomly assigned to LP and LP-losartan groups (AT1 receptor blockade: 10 mg/kg/day until 20 weeks of age). At 20 weeks of age, blood pressure levels were higher and renal RAS was activated in the LP group. We also observed several adverse effects in the kidneys of the LP group, including a higher number of CD3, CD68 and proliferating cell nuclear antigen-positive cells and higher levels of collagen and reactive oxygen species in the kidney. Further, our results revealed that post-weaning losartan treatment completely abolished immune cell infiltration and intrarenal RAS activation in the kidneys of LP rats. The prevention of augmentation of angiotensin (Ang II) concentration abolished inflammatory and fibrotic events, indicating that Ang II via the AT1 receptor is essential for pathological initiation. Our results suggest that the prenatal programming of hypertension is dependent on the up-regulation of local RAS and presence of immune cells in the kidney.
Collapse
|
29
|
Baum M. Role of renal sympathetic nerve activity in prenatal programming of hypertension. Pediatr Nephrol 2018; 33:409-419. [PMID: 27001053 DOI: 10.1007/s00467-016-3359-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 12/30/2022]
Abstract
Prenatal insults, such as maternal dietary protein deprivation and uteroplacental insufficiency, lead to small for gestational age (SGA) neonates. Epidemiological studies from many different parts of the world have shown that SGA neonates are at increased risk for hypertension and early death from cardiovascular disease as adults. Animal models, including prenatal administration of dexamethasone, uterine artery ligation and maternal dietary protein restriction, result in SGA neonates with fewer nephrons than controls. These models are discussed in this educational review, which provides evidence that prenatal insults lead to altered sodium transport in multiple nephron segments. The factors that could result in increased sodium transport are discussed, focusing on new information that there is increased renal sympathetic nerve activity that may be responsible for augmented renal tubular sodium transport. Renal denervation abrogates the hypertension in programmed rats but has no effect on control rats. Other potential factors that could cause hypertension in programmed rats, such as the renin-angiotensin system, are also discussed.
Collapse
Affiliation(s)
- Michel Baum
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Building, Dallas, TX, 75390-9063, USA. .,Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
30
|
Logue OC, Mahdi F, Chapman H, George EM, Bidwell GL. A Maternally Sequestered, Biopolymer-Stabilized Vascular Endothelial Growth Factor (VEGF) Chimera for Treatment of Preeclampsia. J Am Heart Assoc 2017; 6:e007216. [PMID: 29629873 PMCID: PMC5779036 DOI: 10.1161/jaha.117.007216] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preeclampsia is a hypertensive syndrome that complicates 3% to 5% of pregnancies in the United States. Preeclampsia originates from an improperly vascularized and ischemic placenta that releases factors that drive systemic pathophysiology. One of these factors, soluble fms-like tyrosine kinase-1, is believed to sequester vascular endothelial growth factor (VEGF), leading to systemic endothelial dysfunction and hypertension. With the goal of targeting soluble fms-like tyrosine kinase-1 while simultaneously preventing fetal exposure to VEGF, we fused VEGF to elastin-like polypeptide, a biopolymer carrier that does not cross the placental barrier (ELP-VEGF). METHODS AND RESULTS ELP-VEGF restored in vitro endothelial cell tube formation in the presence of plasma from placental ischemic rats. Long-term administered ELP-VEGF in pregnant rats accumulated in maternal kidneys, aorta, liver, and placenta, but the protein was undetectable in the pups when administered at therapeutic doses in dams. Long-term administration of ELP-VEGF in a placental ischemia rat model achieved dose-dependent attenuation of hypertension, with blood pressure equal to sham controls at a dose of 5 mg/kg per day. ELP-VEGF infusion increased total plasma soluble fms-like tyrosine kinase-1 levels but dramatically reduced free plasma soluble fms-like tyrosine kinase-1 and induced urinary excretion of nitrate/nitrite, indicating enhanced renal nitric oxide signaling. ELP-VEGF at up to 5 mg/kg per day had no deleterious effect on maternal or fetal body weight. However, dose-dependent adverse events were observed, including ascites production and neovascular tissue encapsulation around the minipump. CONCLUSIONS ELP-VEGF has the potential to treat the preeclampsia maternal syndrome, but careful dosing and optimization of the delivery route are necessary.
Collapse
Affiliation(s)
- Omar C Logue
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Fakhri Mahdi
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Heather Chapman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
31
|
Prenatal hypoxia leads to hypertension, renal renin-angiotensin system activation and exacerbates salt-induced pathology in a sex-specific manner. Sci Rep 2017; 7:8241. [PMID: 28811528 PMCID: PMC5557956 DOI: 10.1038/s41598-017-08365-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/10/2017] [Indexed: 01/05/2023] Open
Abstract
Prenatal hypoxia is associated with growth restriction and adverse cardiovascular outcomes. Here, we describe renal and cardiovascular outcomes in ageing mouse offspring prenatally exposed to hypoxia (12% O2) from embryonic day 14.5 until birth. At 12 months of age, both male and female offspring exposed to prenatal hypoxia had elevated mean arterial pressure. Glomerular number was reduced by 25% in hypoxia-exposed male, but not female, offspring and this was associated with increased urinary albumin excretion, glomerular hypertrophy and renal fibrosis. Hypoxia-exposed offspring of both sexes were more susceptible to salt-induced cardiac fibrosis, however, renal fibrosis was exacerbated by high salt in males only. In male but not female hypoxia-exposed offspring, renal renin mRNA was increased at weaning. By 12 months, renal renin mRNA expression and concentrations were elevated in both sexes. mRNA expression of At1aR was also elevated in male hypoxia-exposed offspring at 12 months. These results demonstrate that prenatal hypoxia programs elevated blood pressure and exacerbates salt-induced cardiovascular and renal pathology in a sex specific manner. Given sex differences observed in RAS expression and nephron number, future studies may consider RAS blockade as a therapeutic target in this model.
Collapse
|
32
|
Brophy P. Maternal determinants of renal mass and function in the fetus and neonate. Semin Fetal Neonatal Med 2017; 22:67-70. [PMID: 28347404 DOI: 10.1016/j.siny.2017.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The impact of adverse maternal and early gestational issues, ranging from maternal-fetal interactions all the way through to premature birth, are recognized as having influence on the subsequent development of chronic diseases later in life. The development of chronic kidney disease (CKD) as a direct result of early life renal injury or a sequela of diseases such as hypertension or diabetes is a good model example of the potential impact that early life events may have on renal development and lifelong function. The global monetary and human resource cost of CKD is exorbitant. Socio-economic factors, along with other factors (genetic and environmental) may significantly influence the timing and display of phenotypic expression in fetuses and neonates at risk for developing CKD, yet very few of these factors are studied or well understood. In general our focus has been directed at treatment once CKD is established. This strategy has been and remains short-sighted and costly. Earlier understanding of the intrauterine determinants of renal mass development (i.e. environmental "biomes", poor maternal-fetal health, socio-economic factors impacting early life events, diet, access to value based health care and educational opportunities on disease evolution) may allow us an opportunity for earlier intervention. This article aims to provide some foundation for improved understanding of the maternal determinants of renal mass and function in the fetus and neonate.
Collapse
Affiliation(s)
- Patrick Brophy
- University of Iowa Stead Family Children's Hospital, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
33
|
Dasinger JH, Intapad S, Backstrom MA, Carter AJ, Alexander BT. Intrauterine growth restriction programs an accelerated age-related increase in cardiovascular risk in male offspring. Am J Physiol Renal Physiol 2016; 311:F312-9. [PMID: 27147668 PMCID: PMC5005278 DOI: 10.1152/ajprenal.00123.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/27/2016] [Indexed: 11/22/2022] Open
Abstract
Placental insufficiency programs an increase in blood pressure associated with a twofold increase in serum testosterone in male growth-restricted offspring at 4 mo of age. Population studies indicate that the inverse relationship between birth weight and blood pressure is amplified with age. Thus, we tested the hypothesis that intrauterine growth restriction programs an age-related increase in blood pressure in male offspring. Growth-restricted offspring retained a significantly higher blood pressure at 12 but not at 18 mo of age compared with age-matched controls. Blood pressure was significantly increased in control offspring at 18 mo of age relative to control counterparts at 12 mo; however, blood pressure was not increased in growth-restricted at 18 mo relative to growth-restricted counterparts at 12 mo. Serum testosterone levels were not elevated in growth-restricted offspring relative to control at 12 mo of age. Thus, male growth-restricted offspring no longer exhibited a positive association between blood pressure and testosterone at 12 mo of age. Unlike hypertension in male growth-restricted offspring at 4 mo of age, inhibition of the renin-angiotensin system with enalapril (250 mg/l for 2 wk) did not abolish the difference in blood pressure in growth-restricted offspring relative to control counterparts at 12 mo of age. Therefore, these data suggest that intrauterine growth restriction programs an accelerated age-related increase in blood pressure in growth-restricted offspring. Furthermore, this study suggests that the etiology of increased blood pressure in male growth-restricted offspring at 12 mo of age differs from that at 4 mo of age.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Suttira Intapad
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Miles A Backstrom
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Anthony J Carter
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
34
|
Abstract
Hypertension is a risk factor for cardiovascular disease, the leading cause of death worldwide. Although multiple factors contribute to the pathogenesis of hypertension, studies by Dr David Barker reporting an inverse relationship between birth weight and blood pressure led to the hypothesis that slow growth during fetal life increased blood pressure and the risk for cardiovascular disease in later life. It is now recognized that growth during infancy and childhood, in addition to exposure to adverse influences during fetal life, contributes to the developmental programming of increased cardiovascular risk. Numerous epidemiological studies support the link between influences during early life and later cardiovascular health; experimental models provide proof of principle and indicate that numerous mechanisms contribute to the developmental origins of chronic disease. Sex has an impact on the severity of cardiovascular risk in experimental models of developmental insult. Yet, few studies examine the influence of sex on blood pressure and cardiovascular health in low-birth weight men and women. Fewer still assess the impact of ageing on sex differences in programmed cardiovascular risk. Thus, the aim of the present review is to highlight current data about sex differences in the developmental programming of blood pressure and cardiovascular disease.
Collapse
|
35
|
Perrone S, Santacroce A, Picardi A, Buonocore G. Fetal programming and early identification of newborns at high risk of free radical-mediated diseases. World J Clin Pediatr 2016; 5:172-181. [PMID: 27170927 PMCID: PMC4857230 DOI: 10.5409/wjcp.v5.i2.172] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/30/2015] [Accepted: 02/16/2016] [Indexed: 02/06/2023] Open
Abstract
Nowadays metabolic syndrome represents a real outbreak affecting society. Paradoxically, pediatricians must feel involved in fighting this condition because of the latest evidences of developmental origins of adult diseases. Fetal programming occurs when the normal fetal development is disrupted by an abnormal insult applied to a critical point in intrauterine life. Placenta assumes a pivotal role in programming the fetal experience in utero due to the adaptive changes in structure and function. Pregnancy complications such as diabetes, intrauterine growth restriction, pre-eclampsia, and hypoxia are associated with placental dysfunction and programming. Many experimental studies have been conducted to explain the phenotypic consequences of fetal-placental perturbations that predispose to the genesis of metabolic syndrome, obesity, diabetes, hyperinsulinemia, hypertension, and cardiovascular disease in adulthood. In recent years, elucidating the mechanisms involved in such kind of process has become the challenge of scientific research. Oxidative stress may be the general underlying mechanism that links altered placental function to fetal programming. Maternal diabetes, prenatal hypoxic/ischaemic events, inflammatory/infective insults are specific triggers for an acute increase in free radicals generation. Early identification of fetuses and newborns at high risk of oxidative damage may be crucial to decrease infant and adult morbidity.
Collapse
|
36
|
Morton JS, Cooke CL, Davidge ST. In Utero Origins of Hypertension: Mechanisms and Targets for Therapy. Physiol Rev 2016; 96:549-603. [DOI: 10.1152/physrev.00015.2015] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The developmental origins of health and disease theory is based on evidence that a suboptimal environment during fetal and neonatal development can significantly impact the evolution of adult-onset disease. Abundant evidence exists that a compromised prenatal (and early postnatal) environment leads to an increased risk of hypertension later in life. Hypertension is a silent, chronic, and progressive disease defined by elevated blood pressure (>140/90 mmHg) and is strongly correlated with cardiovascular morbidity/mortality. The pathophysiological mechanisms, however, are complex and poorly understood, and hypertension continues to be one of the most resilient health problems in modern society. Research into the programming of hypertension has proposed pharmacological treatment strategies to reverse and/or prevent disease. In addition, modifications to the lifestyle of pregnant women might impart far-reaching benefits to the health of their children. As more information is discovered, more successful management of hypertension can be expected to follow; however, while pregnancy complications such as fetal growth restriction, preeclampsia, preterm birth, etc., continue to occur, their offspring will be at increased risk for hypertension. This article reviews the current knowledge surrounding the developmental origins of hypertension, with a focus on mechanistic pathways and targets for therapeutic and pharmacologic interventions.
Collapse
Affiliation(s)
- Jude S. Morton
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| | - Christy-Lynn Cooke
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| | - Sandra T. Davidge
- Departments of Obstetrics and Gynaecology and of Physiology, University of Alberta, Edmonton, Canada; Women and Children's Health Research Institute, Edmonton, Canada; and Cardiovascular Research Centre, Edmonton, Canada
| |
Collapse
|
37
|
Mansuri A, Elmaghrabi A, Legan SK, Gattineni J, Baum M. Transient Exposure of Enalapril Normalizes Prenatal Programming of Hypertension and Urinary Angiotensinogen Excretion. PLoS One 2015; 10:e0146183. [PMID: 26719973 PMCID: PMC4699824 DOI: 10.1371/journal.pone.0146183] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Maternal low protein diet programs offspring to develop hypertension as adults. Transient exposure to angiotensin converting enzyme inhibitors or angiotensin II receptor blockers can result in improvement in hypertension. Male rats whose mothers received a low protein diet during the last half of pregnancy were given either vehicle, continuous enalapril (CE) in their drinking water or were given transient enalapril exposure (TE) after weaning at 21 days of age. The TE group had enalapril in their drinking water for 21 days starting from day 21 of life. All rats were studied at 6 months of age. Vehicle treated rats whose mothers were fed a low protein diet were hypertensive, had albuminuria, and demonstrated upregulation of the intrarenal renin-angiotensin system as evidenced by higher urinary angiotensinogen and urinary angiotensin II levels. In low protein rats both continuous and transient exposure to enalapril normalized blood pressure, urinary angiotensinogen and urinary angiotensin II levels at 6 months of age, but only continuous administration of enalapril decreased urinary albumin excretion. These data support the importance of the intrarenal renin-angiotensin system in mediating hypertension in programmed rats and transient exposure to enalapril can reprogram the hypertension and dysregulation of the intrarenal renin-angiotensin system.
Collapse
Affiliation(s)
- Asifhusen Mansuri
- Department of Pediatrics University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ayah Elmaghrabi
- Department of Pediatrics University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Susan K. Legan
- Department of Pediatrics University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jyothsna Gattineni
- Department of Pediatrics University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michel Baum
- Department of Pediatrics University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Internal Medicine University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Lino CA, da Silva IB, Shibata CER, Monteiro PDS, Barreto-Chaves MLM. Maternal hyperthyroidism increases the susceptibility of rat adult offspring to cardiovascular disorders. Mol Cell Endocrinol 2015; 416:1-8. [PMID: 26277399 DOI: 10.1016/j.mce.2015.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022]
Abstract
Suboptimal intrauterine conditions as changed hormone levels during critical periods of the development are considered an insult and implicate in physiological adaptations which may result in pathological outcomes in later life. This study evaluated the effect of maternal hyperthyroidism (hyper) on cardiac function in adult offspring and the possible involvement of cardiac Renin-Angiotensin System (RAS) in this process. Wistar dams received orally thyroxin (12 mg/L) from gestational day 9 (GD9) until GD18. Adult offspring at postnatal day 90 (PND90) from hyper dams presented increased SBP evaluated by plethysmography and worse recovery after ischemia-reperfusion (I/R), as evidenced by decreased LVDP, +dP/dT and -dP/dT at 25 min of reperfusion and by increased infarct size. Increased cardiac Angiotensin I/II levels and AT1R in hyper offspring were verified. Herein, we provide evidences that maternal hyperthyroidism leads to altered expression of RAS components in adult offspring, which may be correlated with worse recovery of the cardiac performance after ischemic insults and hypertension.
Collapse
Affiliation(s)
- Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ivson Bezerra da Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Caroline E R Shibata
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Priscilla de S Monteiro
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
39
|
Intapad S, Ojeda NB, Varney E, Royals TP, Alexander BT. Sex-Specific Effect of Endothelin in the Blood Pressure Response to Acute Angiotensin II in Growth-Restricted Rats. Hypertension 2015; 66:1260-6. [PMID: 26459423 DOI: 10.1161/hypertensionaha.115.06257] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/21/2015] [Indexed: 01/18/2023]
Abstract
The renal endothelin system contributes to sex differences in blood pressure with males demonstrating greater endothelin type-A receptor-mediated responses relative to females. Intrauterine growth restriction programs hypertension and enhance renal sensitivity to acute angiotensin II in male growth-restricted rats. Endothelin is reported to work synergistically with angiotensin II. Thus, this study tested the hypothesis that endothelin augments the blood pressure response to acute angiotensin II in male growth-restricted rats. Systemic and renal hemodynamics were determined in response to acute angiotensin II (100 mg/kg per minute for 30 minutes) with and without the endothelin type-A receptor antagonist, Atrasentan (ABT-627; 10 ng/kg per minute for 30 minutes), in rats pretreated with enalapril (250 mg/L for 1 week) to normalize the endogenous renin-angiotensin system. Endothelin type-A receptor blockade reduced angiotensin II-mediated increases in blood pressure in male control and male growth-restricted rats. Endothelin type-A receptor blockade also abolished hyper-responsiveness to acute angiotensin II in male growth-restricted rats. Yet, blood pressure remained significantly elevated above baseline after endothelin type-A receptor blockade, suggesting that factors in addition to endothelin contribute to the basic angiotensin II-induced pressor response in male rats. We also determined sex-specific effects of endothelin on acute angiotensin II-mediated hemodynamic responses. Endothelin type-A receptor blockade did not reduce acute angiotensin II-mediated increases in blood pressure in female control or growth-restricted rats, intact or ovariectomized. Thus, these data suggest that endothelin type-A receptor blockade contributes to hypersensitivity to acute angiotensin II in male growth-restricted rats and further supports the sex-specific effect of endothelin on blood pressure.
Collapse
Affiliation(s)
- Suttira Intapad
- Department of Pediatrics (N.B.O.), Department of Physiology and Biophysics (S.I., N.B.O., E.V., T.P.R., B.T.A.), and the Center for Developmental Disorders Research (S.I., N.B.O., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Norma B Ojeda
- Department of Pediatrics (N.B.O.), Department of Physiology and Biophysics (S.I., N.B.O., E.V., T.P.R., B.T.A.), and the Center for Developmental Disorders Research (S.I., N.B.O., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Elliott Varney
- Department of Pediatrics (N.B.O.), Department of Physiology and Biophysics (S.I., N.B.O., E.V., T.P.R., B.T.A.), and the Center for Developmental Disorders Research (S.I., N.B.O., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Thomas P Royals
- Department of Pediatrics (N.B.O.), Department of Physiology and Biophysics (S.I., N.B.O., E.V., T.P.R., B.T.A.), and the Center for Developmental Disorders Research (S.I., N.B.O., B.T.A.), University of Mississippi Medical Center, Jackson
| | - Barbara T Alexander
- Department of Pediatrics (N.B.O.), Department of Physiology and Biophysics (S.I., N.B.O., E.V., T.P.R., B.T.A.), and the Center for Developmental Disorders Research (S.I., N.B.O., B.T.A.), University of Mississippi Medical Center, Jackson.
| |
Collapse
|
40
|
Ouabain rescues rat nephrogenesis during intrauterine growth restriction by regulating the complement and coagulation cascades and calcium signaling pathway. J Dev Orig Health Dis 2015; 7:91-101. [PMID: 26442628 DOI: 10.1017/s2040174415007242] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrauterine growth restriction (IUGR) is associated with a reduction in the numbers of nephrons in neonates, which increases the risk of hypertension. Our previous study showed that ouabain protects the development of the embryonic kidney during IUGR. To explore this molecular mechanism, IUGR rats were induced by protein and calorie restriction throughout pregnancy, and ouabain was delivered using a mini osmotic pump. RNA sequencing technology was used to identify the differentially expressed genes (DEGs) of the embryonic kidneys. DEGs were submitted to the Database for Annotation and Visualization and Integrated Discovery, and gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were conducted. Maternal malnutrition significantly reduced fetal weight, but ouabain treatment had no significant effect on body weight. A total of 322 (177 upregulated and 145 downregulated) DEGs were detected between control and the IUGR group. Meanwhile, 318 DEGs were found to be differentially expressed (180 increased and 138 decreased) between the IUGR group and the ouabain-treated group. KEGG pathway analysis indicated that maternal undernutrition mainly disrupts the complement and coagulation cascades and the calcium signaling pathway, which could be protected by ouabain treatment. Taken together, these two biological pathways may play an important role in nephrogenesis, indicating potential novel therapeutic targets against the unfavorable effects of IUGR.
Collapse
|
41
|
Abstract
Low birth weight serves as a crude proxy for impaired growth during fetal life and indicates a failure for the fetus to achieve its full growth potential. Low birth weight can occur in response to numerous etiologies that include complications during pregnancy, poor prenatal care, parental smoking, maternal alcohol consumption, or stress. Numerous epidemiological and experimental studies demonstrate that birth weight is inversely associated with blood pressure and coronary heart disease. Sex and age impact the developmental programming of hypertension. In addition, impaired growth during fetal life also programs enhanced vulnerability to a secondary insult. Macrosomia, which occurs in response to maternal obesity, diabetes, and excessive weight gain during gestation, is also associated with increased cardiovascular risk. Yet, the exact mechanisms that permanently change the structure, physiology, and endocrine health of an individual across their lifespan following altered growth during fetal life are not entirely clear. Transmission of increased risk from one generation to the next in the absence of an additional prenatal insult indicates an important role for epigenetic processes. Experimental studies also indicate that the sympathetic nervous system, the renin angiotensin system, increased production of oxidative stress, and increased endothelin play an important role in the developmental programming of blood pressure in later life. Thus, this review will highlight how adverse influences during fetal life and early development program an increased risk for cardiovascular disease including high blood pressure and provide an overview of the underlying mechanisms that contribute to the fetal origins of cardiovascular pathology.
Collapse
Affiliation(s)
- Barbara T Alexander
- Department of Physiology and Biophysics, Women's Health Research Center, Center for Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | | |
Collapse
|
42
|
Wang KCW, Brooks DA, Summers-Pearce B, Bobrovskaya L, Tosh DN, Duffield JA, Botting KJ, Zhang S, Caroline McMillen I, Morrison JL. Low birth weight activates the renin-angiotensin system, but limits cardiac angiogenesis in early postnatal life. Physiol Rep 2015; 3:3/2/e12270. [PMID: 25649246 PMCID: PMC4393187 DOI: 10.14814/phy2.12270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Low birth weight (LBW) is associated with increased risk of adult cardiovascular disease and this association may be partly a consequence of early programming of the renin-angiotensin system (RAS). We investigated the effects of LBW on expression of molecules in the RAS and cardiac tissue remodeling. Left ventricular samples were collected from the hearts of 21 days old lambs that were born average birth weight (ABW) and LBW. Cardiac mRNA expression was quantified using real-time RT-PCR and protein expression was quantified using Western blotting. DNA methylation and histone acetylation were assessed by combined bisulfite restriction analysis and chromatin immunoprecipitation, respectively. There were increased plasma renin activity, angiotensin I (ANGI), and ANGII concentrations in LBW compared to ABW lambs at day 20. In LBW lambs, there was increased expression of cardiac ACE2 mRNA, decreased ANGII receptor type 1 (AT1R) protein, and acetylation of histone H3K9 of the AT1R promoter but no changes in AT1R mRNA expression and AT1R promoter DNA methylation. There was no difference in the abundance of proteins involved in autophagy or fibrosis. BIRC5 and VEGF mRNA expression was increased; however, the total length of the capillaries was decreased in the hearts of LBW lambs. Activation of the circulating and local cardiac RAS in neonatal LBW lambs may be expected to increase cardiac fibrosis, autophagy, and capillary length. However, we observed only a decrease in total capillary length, suggesting a dysregulation of the RAS in the heart of LBW lambs and this may have significant implications for heart health in later life.
Collapse
Affiliation(s)
- Kimberley C W Wang
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Brooke Summers-Pearce
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Larisa Bobrovskaya
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Darran N Tosh
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Jaime A Duffield
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Song Zhang
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
43
|
Intapad S, Ojeda NB, Dasinger JH, Alexander BT. Sex differences in the developmental origins of cardiovascular disease. Physiology (Bethesda) 2014; 29:122-32. [PMID: 24583768 DOI: 10.1152/physiol.00045.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The Developmental Origins of Health and Disease (DOHaD) proposes that adverse events during early life program an increased risk for cardiovascular disease. Experimental models provide proof of concept but also indicate that insults during early life program sex differences in adult blood pressure and cardiovascular risk. This review will highlight the potential mechanisms that contribute to the etiology of sex differences in the developmental programming of cardiovascular disease.
Collapse
Affiliation(s)
- Suttira Intapad
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | | |
Collapse
|
44
|
Gois PHF, Canale D, Luchi WM, Volpini RA, Veras MM, Costa NDSX, Shimizu MHM, Seguro AC. Tenofovir during pregnancy in rats: a novel pathway for programmed hypertension in the offspring. J Antimicrob Chemother 2014; 70:1094-105. [PMID: 25492393 DOI: 10.1093/jac/dku483] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To evaluate the occurrence of systemic and renal abnormalities in the offspring of Wistar rats exposed to tenofovir disoproxil fumarate (DF) during pregnancy. METHODS Female Wistar rats received a standard diet, with or without addition of tenofovir DF (100 mg/kg diet), 1 week before mating and during pregnancy. Offspring from the tenofovir DF group were placed with an untreated foster mother during breastfeeding and compared with offspring from rats maintained on a standard diet during mating and pregnancy (control). Control and tenofovir DF were followed up at 3 and 6 months of age. Monthly body weight and systolic blood pressure (SBP), glomerular counts, renal function, biochemical parameters, angiotensin II, renal renin angiotensin aldosterone system (RAAS) and renal sodium transporters were analysed. RESULTS Tenofovir DF offspring showed lower birth weight compared with the control group. After the third month, growth among the tenofovir DF group experienced a rapid catch-up. SBP increased progressively after the second month of age in the tenofovir DF group. Nephron number did not differ between the groups; however, the tenofovir DF group showed glomerular structural changes. Plasma aldosterone was higher in the tenofovir DF group, associated with a significant increase in renal expression of RAAS. The tenofovir DF rats showed up-regulation of renal sodium transporters and consequently lower urinary sodium excretion. CONCLUSIONS This is the first demonstration using an experimental model that maternal exposure to tenofovir DF during gestation results in overactivation of RAAS, up-regulation of renal sodium transporters and hypertension in the offspring.
Collapse
Affiliation(s)
- Pedro Henrique França Gois
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Daniele Canale
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Weverton Machado Luchi
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Rildo Aparecido Volpini
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Mariana Matera Veras
- Laboratory of Experimental Air Pollution-LIM05, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Natália de Souza Xavier Costa
- Laboratory of Experimental Air Pollution-LIM05, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Maria Heloisa Massola Shimizu
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Antonio Carlos Seguro
- Laboratory for Medical Research-LIM12, Nephrology Department, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
45
|
Saez F, Reverte V, Paliege A, Moreno JM, Llinás MT, Bachmann S, Salazar FJ. Sex-dependent hypertension and renal changes in aged rats with altered renal development. Am J Physiol Renal Physiol 2014; 307:F461-70. [DOI: 10.1152/ajprenal.00198.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Numerous studies have evaluated blood pressure (BP) and renal changes in several models of developmental programming of hypertension. The present study examined to what extent BP, renal hemodynamic, and renal structure are affected at an old age in male and female animals with altered renal development. It also evaluated whether renal damage is associated with changes in cyclooxygenase (COX)-2 and neuronal nitric oxide synthase (NOS1) expression and immunoreactivity. Experiments were carried out in rats at 10–11 and 16–17 mo of age treated with vehicle or an ANG II type 1 receptor antagonist during the nephrogenic period (ARAnp). A progressive increment in BP and a deterioration of renal hemodynamics were found in both sexes of ARAnp-treated rats, with these changes being greater ( P < 0.05) in male rats. The decrease in glomerular filtration rate at the oldest age was greater ( P < 0.05) in male (74%) than female (32%) ARAnp-treated rats. Sex-dependent deterioration of renal structure was demonstrated in optical and electron microscopic experiments. COX-2 and NOS1 immunoreactivity were enhanced in the macula densa of male but not female ARAnp-treated rats. The present study reports novel findings suggesting that stimuli that induce a decrease of ANG II effects during renal development lead to a progressive increment in BP and renal damage at an old age in both sexes, but these BP and renal changes are greater in males than in females. The renal damage is associated with an increase of COX-2 and NOS1 in the macula densa of males but not females with altered renal development.
Collapse
Affiliation(s)
- Fara Saez
- Department of Physiology, University of Murcia, Regional Campus of International Excellence “Mare Nostrum,” Murcia, Instituto Murciano de Investigación Biomédica, Spain; and
| | - Virginia Reverte
- Department of Physiology, University of Murcia, Regional Campus of International Excellence “Mare Nostrum,” Murcia, Instituto Murciano de Investigación Biomédica, Spain; and
| | - Alexander Paliege
- Anatomisches Institut, Charité, Humboldt Universität, Berlin, Germany
| | - Juan Manuel Moreno
- Department of Physiology, University of Murcia, Regional Campus of International Excellence “Mare Nostrum,” Murcia, Instituto Murciano de Investigación Biomédica, Spain; and
| | - María T. Llinás
- Department of Physiology, University of Murcia, Regional Campus of International Excellence “Mare Nostrum,” Murcia, Instituto Murciano de Investigación Biomédica, Spain; and
| | | | - F. Javier Salazar
- Department of Physiology, University of Murcia, Regional Campus of International Excellence “Mare Nostrum,” Murcia, Instituto Murciano de Investigación Biomédica, Spain; and
| |
Collapse
|
46
|
Kooijman MN, Bakker H, van der Heijden AJ, Hofman A, Franco OH, Steegers EAP, Taal HR, Jaddoe VWV. Childhood kidney outcomes in relation to fetal blood flow and kidney size. J Am Soc Nephrol 2014; 25:2616-24. [PMID: 24812167 DOI: 10.1681/asn.2013070746] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Impaired fetal abdominal blood flow may lead to smaller kidneys and subsequent impaired kidney function in later life. In a prospective cohort study among 923 pregnant women and their children, we measured fetal growth, kidney volumes, and umbilical and cerebral artery blood flow (median gestational age of 30.3 weeks; 95% range, 28.5-32.7 weeks). We used a higher umbilical/cerebral artery pulsatility index ratio as an indicator of preferential fetal blood flow to the upper body parts at the expense of the intra-abdominal organs. At a median age of 5.9 years (95% range, 5.7-6.6 years), we measured childhood kidney volumes, creatinine and cystatin C blood levels, microalbuminuria, BP, and eGFR. A preferential fetal blood flow to the upper body parts at the expense of the intra-abdominal organs associated only with a smaller combined kidney volume in childhood. Fetal combined kidney volume positively associated with childhood combined kidney volume and eGFR, and inversely associated with childhood creatinine and cystatin C levels (all P values <0.05), but did not associate with childhood microalbuminuria and BP. Children within the highest tertile of fetal umbilical/cerebral ratio and the lowest tertile of fetal combined kidney volume had the lowest eGFR (difference, -6.36 ml/min per 1.73 m(2); 95% confidence interval, -11.78 to -0.94 compared with children within the middle tertiles). These data suggest that impaired fetal blood to the abdominal organs and smaller fetal kidney size are associated with subclinical changes in kidney outcomes in school-aged children.
Collapse
Affiliation(s)
| | - Hanneke Bakker
- The Generation R Study Group, Departments of Epidemiology, Pediatrics, and
| | | | | | | | - Eric A P Steegers
- Gynecology and Obstetrics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Vincent W V Jaddoe
- The Generation R Study Group, Departments of Epidemiology, Pediatrics, and
| |
Collapse
|
47
|
Bakker H, Gaillard R, Franco OH, Hofman A, van der Heijden AJ, Steegers EAP, Taal HR, Jaddoe VWV. Fetal and infant growth patterns and kidney function at school age. J Am Soc Nephrol 2014; 25:2607-15. [PMID: 24812164 DOI: 10.1681/asn.2013091003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Low birth weight is associated with ESRD. To identify specific growth patterns in early life that may be related to kidney function in later life, we examined the associations of longitudinally measured fetal and infant growth with kidney function in school-aged children. This study was embedded in a population-based prospective cohort study among 6482 children followed from fetal life onward. Fetal and childhood growth was measured during second and third trimesters of pregnancy, at birth, and at 6, 12, 24, 36, and 48 months postnatally. At the age of 6 years, we measured kidney volume by ultrasound. GFR was estimated using blood creatinine levels. Higher gestational age-adjusted birth weight was associated with higher combined kidney volume and higher eGFR (per 1 SD score increase in birth weight; 1.27 cm(3) [95% confidence interval, 0.61 to 1.93] and 0.78 ml/min per 1.73 m2 [95% CI, 0.16 to 1.39], respectively). Fetal weight, birth weight, and weight at 6 months were positively associated with childhood kidney volume, whereas higher second trimester fetal weight was positively associated with higher GFR (all P values<0.05). Fetal and childhood lengths were not consistently associated with kidney function. In this cohort, lower fetal and early infant weight growth is associated with smaller kidney volume in childhood, whereas only lower fetal weight growth is associated with lower kidney function in childhood, independent of childhood growth. Whether these associations lead to an increased risk of kidney disease needs to be studied further.
Collapse
Affiliation(s)
- Hanneke Bakker
- The Generation R Study Group, and Departments of Epidemiology, Pediatrics, and
| | - Romy Gaillard
- The Generation R Study Group, and Departments of Epidemiology, Pediatrics, and
| | | | | | | | - Eric A P Steegers
- Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Vincent W V Jaddoe
- The Generation R Study Group, and Departments of Epidemiology, Pediatrics, and
| |
Collapse
|
48
|
Ojeda NB, Intapad S, Alexander BT. Sex differences in the developmental programming of hypertension. Acta Physiol (Oxf) 2014; 210:307-16. [PMID: 24268043 DOI: 10.1111/apha.12206] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 07/29/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
Experimental models of developmental programming provide proof of concept and support Barker's original findings that link birthweight and blood pressure. Many experimental models of developmental insult demonstrate a sex difference with male offspring exhibiting a higher blood pressure in young adulthood relative to their age-matched female counterparts. It is well recognized that men exhibit a higher blood pressure relative to age-matched women prior to menopause. Yet, whether this sex difference is noted in individuals born with low birthweight is not clear. Sex differences in the developmental programming of blood pressure may originate from innate sex-specific differences in expression of the renin angiotensin system that occur in response to adverse influences during early life. Sex differences in the developmental programming of blood pressure may also involve the influence of the hormonal milieu on regulatory systems key to the long-term control of blood pressure such as the renin angiotensin system in adulthood. In addition, the sex difference in blood pressure in offspring exposed to a developmental insult may involve innate sex differences in oxidative status or the endothelin system or may be influenced by age-dependent changes in the developmental programming of cardiovascular risk factors such as adiposity. Therefore, this review will highlight findings from different experimental models to provide the current state of knowledge related to the mechanisms that contribute to the aetiology of sex differences in the developmental programming of blood pressure and hypertension.
Collapse
Affiliation(s)
- N. B. Ojeda
- Department of Pediatrics; University of Mississippi Medical Center; Jackson MS USA
- Women's Health Research Center; University of Mississippi Medical Center; Jackson MS USA
| | - S. Intapad
- Department of Physiology and Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - B. T. Alexander
- Women's Health Research Center; University of Mississippi Medical Center; Jackson MS USA
- Department of Physiology and Biophysics; University of Mississippi Medical Center; Jackson MS USA
| |
Collapse
|
49
|
Gallo LA, Tran M, Cullen-McEwen LA, Denton KM, Jefferies AJ, Moritz KM, Wlodek ME. Transgenerational programming of fetal nephron deficits and sex-specific adult hypertension in rats. Reprod Fertil Dev 2014; 26:1032-43. [DOI: 10.1071/rd13133] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/02/2013] [Indexed: 01/27/2023] Open
Abstract
A developmental insult that restricts growth in the first generation has the potential to program disease in subsequent generations. The aim of this study was to ascertain transgenerational growth and cardio–renal effects, via the maternal line, in a rat model of utero–placental insufficiency. Bilateral uterine vessel ligation or sham surgery (offspring termed first generation; F1 Restricted and Control, respectively) was performed in WKY rats. F1 Restricted and Control females were mated with normal males to produce second generation (F2) offspring (Restricted and Control) studied from fetal (embryonic Day 20) to adult (12 months) life. F2 Restricted male and female fetuses had reduced (P < 0.05) nephron number (down 15–22%) but this deficit was not sustained postnatally and levels were similar to Controls at Day 35. F2 Restricted males, but not females, developed elevated (+16 mmHg, P < 0.05) systolic blood pressure at 6 months of age, which was sustained to 9 months. This was not explained by alterations to intra-renal or plasma components of the renin–angiotensin system. In a rat model of utero–placental insufficiency, we report alterations to F2 kidney development and sex-specific adult hypertension. This study demonstrates that low birthweight can have far-reaching effects that extend into the next generation.
Collapse
|
50
|
Paixão AD, Alexander BT. How the kidney is impacted by the perinatal maternal environment to develop hypertension. Biol Reprod 2013; 89:144. [PMID: 24227755 DOI: 10.1095/biolreprod.113.111823] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Environmental conditions during perinatal development such as maternal undernutrition, maternal glucocorticoids, placental insufficiency, and maternal sodium overload can program changes in renal Na(+) excretion leading to hypertension. Experimental studies indicate that fetal exposure to an adverse maternal environment may reduce glomerular filtration rate by decreasing the surface area of the glomerular capillaries. Moreover, fetal responses to environmental insults during early life that contribute to the development of hypertension may include increased expression of tubular apical or basolateral membrane Na(+) transporters and increased production of renal superoxide leading to enhanced Na(+) reabsorption. This review will address the role of these potential renal mechanisms in the fetal programming of hypertension in experimental models induced by maternal undernutrition, fetal exposure to glucocorticoids, placental insufficiency, and maternal sodium overload in the rat.
Collapse
Affiliation(s)
- Ana D Paixão
- Department of Physiology and Pharmacology, Center of Biological Sciences, Federal University of Pernambuco, Recife, Brazil
| | | |
Collapse
|