1
|
Oulerich Z, Sferruzzi-Perri AN. Early-life exposures and long-term health: adverse gestational environments and the programming of offspring renal and vascular disease. Am J Physiol Renal Physiol 2024; 327:F21-F36. [PMID: 38695077 PMCID: PMC11687964 DOI: 10.1152/ajprenal.00383.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 06/21/2024] Open
Abstract
According to the Developmental Origins of Health and Disease hypothesis, exposure to certain environmental influences during early life may be a key determinant of fetal development and short- and long-term offspring health. Indeed, adverse conditions encountered during the fetal, perinatal, and early childhood stages can alter normal development and growth, as well as put the offspring at elevated risk of developing long-term health conditions in adulthood, including chronic kidney disease and cardiovascular diseases. Of relevance in understanding the mechanistic basis of these long-term health conditions are previous findings showing low glomerular number in human intrauterine growth restriction and low birth weight-indicators of a suboptimal intrauterine environment. In different animal models, the main suboptimal intrauterine conditions studied relate to maternal dietary manipulations, poor micronutrient intake, prenatal ethanol exposure, maternal diabetes, glucocorticoid and chemical exposure, hypoxia, and placental insufficiency. These studies have demonstrated changes in kidney structure, glomerular endowment, and expression of key genes and signaling pathways controlling endocrine, excretion, and filtration function of the offspring. This review aims to summarize those studies to uncover the effects and mechanisms by which adverse gestational environments impact offspring renal and vascular health in adulthood. This is important for identifying agents and interventions that can prevent and mitigate the long-term consequences of an adverse intrauterine environment on the subsequent generation.NEW & NOTEWORTHY Human data and experimental animal data show that suboptimal environments during fetal development increase the risk of renal and vascular diseases in adult-life. This is related to permanent changes in kidney structure, function, and expression of genes and signaling pathways controlling filtration, excretion, and endocrine function. Uncovering the mechanisms by which offspring renal development and function is impacted is important for identifying ways to mitigate the development of diseases that strain health care services worldwide.
Collapse
Affiliation(s)
- Zoé Oulerich
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Agro Paris Tech, Université Paris-Saclay, Paris, France
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
2
|
Ramirez LA, Mohamed R, Marin T, Brands MW, Snyder E, Sullivan JC. Perinatal intermittent hypoxia increases early susceptibility to ANG II-induced hypertension in adult male but not in female Sprague-Dawley rats. Am J Physiol Renal Physiol 2023; 324:F483-F493. [PMID: 36951371 PMCID: PMC10151053 DOI: 10.1152/ajprenal.00308.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/15/2023] [Accepted: 03/16/2023] [Indexed: 03/24/2023] Open
Abstract
Prenatal, perinatal, and adulthood exposure to chronic intermittent hypoxia (IH) increases blood pressure in rodents. Males exposed to chronic IH have higher blood pressure versus females. However, it is unknown if this same-sex difference exists with acute perinatal IH. We tested the hypothesis that acute perinatal IH increases baseline blood pressure and enhances sensitivity to angiotensin II (ANG II)-induced hypertension in male Sprague-Dawley rats. Male and female pups were randomized to control (room air) or IH (10 min of ∼10% O2 for 3 times/day) for the first 8 days of life. IH decreased oxygen saturation, as confirmed via a pulse oximeter. Pups were weaned at postnatal day 21. Blood pressure was measured via telemetry beginning at 14 wk of age and analyzed separately into light and dark phases to assess circadian rhythm. Osmotic minipumps to deliver ANG II were implanted at 15 wk of age. Perinatal IH exposure did not alter baseline blood pressure. One week of ANG II treatment increased blood pressure in light and dark periods in males exposed to IH versus control; there was no effect in females. Blood pressure among the groups was comparable following 2 wk of ANG II infusion. Perinatal IH did not change the circadian rhythm. Following ANG II treatment, indexes of renal injury were measured. Perinatal IH did not alter kidney size, structure, nephron number, or creatinine clearance. These data indicate that acute perinatal IH enhances early ANG II-induced hypertension in males, independent of nephron loss or decreases in body weight or kidney function.NEW & NOTEWORTHY The impact of acute intermittent hypoxia (IH) in early life on blood pressure in adulthood is unknown. This study used a new model exposing female and male rat pups to acute IH in the first 8 days of life, without exposing the dam. Although baseline blood pressure was not altered in adulthood, IH increased susceptibility to angiotensin II hypertension only in males, supporting increased susceptibility of males exposed to IH to a second cardiovascular stressor.
Collapse
Affiliation(s)
- Lindsey A Ramirez
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Terri Marin
- Department of Nursing Science, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Elizabeth Snyder
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
3
|
Meister TA, Soria R, Dogar A, Messerli FH, Paoloni-Giacobino A, Stenz L, Scherrer U, Sartori C, Rexhaj E. Increased Arterial Responsiveness to Angiotensin II in Mice Conceived by Assisted Reproductive Technologies. Int J Mol Sci 2022; 23:13357. [PMID: 36362144 PMCID: PMC9654033 DOI: 10.3390/ijms232113357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 09/01/2023] Open
Abstract
Since the first report in 1978, the number of individuals conceived by Assisted Reproductive Technologies (ART) has grown incessantly. In parallel, with the recent emergence of possible underlying mechanisms of ART-induced epigenetic changes in the renin-angiotensin system, the cardiovascular repercussions of ART in mice and human offspring (including arterial hypertension, vascular dysfunction, and cardiac remodeling) have become increasingly recognized. Here, we hypothesized that ART may increase arterial responsiveness to angiotensin II (ANG II) by epigenetically modifying the expression of its receptors. To test this hypothesis, we assessed the vasoconstrictor responsiveness to ANG II in isolated aortas from ART and control mice. We also examined ANG II receptor (ATR) type 1 and 2 expression and the promoter methylation of the At1aR, At1bR and At2R genes. We found that the vasoconstrictor response to ANG II was markedly increased in ART mice compared to controls. This exaggerated vasoconstrictor responsiveness in ART mice correlated with a significant increase in the ANG II receptor (ATR) type 1 to ATR type 2 protein expression ratio in the aorta; this was mainly driven by an increase in AT1R expression, and by hypomethylation of two CpG sites located in the At1bR gene promoter leading to increased transcription of the gene. We conclude that in mice, ART increase the vasoconstrictor response to ANG II in the aorta by epigenetically causing an imbalance between the expression of vasoconstrictor (AT1R) and vasodilator (AT2R) ANG II receptors. Unbalanced expression of AT1R and AT2R receptors seems to be a novel mechanism contributing to ART-induced arterial hypertension in mice.
Collapse
Affiliation(s)
- Theo Arthur Meister
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
| | - Rodrigo Soria
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
| | - Afzal Dogar
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
- Tropic Biosciences Ltd., Norwich Research Park Innovation Centre, Norwich NR4 7GJ, UK
| | - Franz H. Messerli
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
| | | | - Ludwig Stenz
- Department of Genetic Medicine and Development, University of Geneva, 1205 Geneva, Switzerland
| | - Urs Scherrer
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
| | - Claudio Sartori
- Department of Internal Medicine, Lausanne University Hospital (CHUV), Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Emrush Rexhaj
- Department of Cardiology and Biomedical Research, Inselspital Bern University Hospital, 3010 Bern, Switzerland
| |
Collapse
|
4
|
Meshchaninov VN, Tsyvian PB, Myakotnykh VS, Kovtun OP, Shcherbakov DL, Blagodareva MS. Ontogenetic Principles of Accelerated Aging and the Prospects for Its Prevention and Treatment. ADVANCES IN GERONTOLOGY 2022. [DOI: 10.1134/s2079057022030080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
5
|
Rudloff S, Bileck A, Janker L, Wanner N, Liaukouskaya N, Lundby C, Huber TB, Gerner C, Huynh-Do U. Dichotomous responses to chronic fetal hypoxia lead to a predetermined aging phenotype. Mol Cell Proteomics 2021; 21:100190. [PMID: 34958949 PMCID: PMC8808178 DOI: 10.1016/j.mcpro.2021.100190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/07/2021] [Accepted: 12/21/2021] [Indexed: 12/29/2022] Open
Abstract
Hypoxia-induced intrauterine growth restriction increases the risk for cardiovascular, renal and other chronic diseases in adults, representing thus a major public health problem. Still, not much is known about the fetal mechanisms that predispose these individuals to disease. Using a previously validated mouse model of fetal hypoxia and bottom-up proteomics we characterize the response of the fetal kidney to chronic hypoxic stress. Fetal kidneys exhibit a dichotomous response to chronic hypoxia, comprising on the one hand cellular adaptations that promote survival (glycolysis, autophagy, and reduced DNA and protein synthesis), but on the other processes that induce a senescence-like phenotype (infiltration of inflammatory cells, DNA damage, and reduced proliferation). Importantly, chronic hypoxia also reduces the expression of the anti-aging proteins klotho and Sirt6, a mechanism that is evolutionary conserved between mice and humans. Taken together, we uncover that predetermined aging during fetal development is a key event in chronic hypoxia, establishing a solid foundation for Barker's hypothesis of fetal programming of adult diseases. This phenotype is associated with a characteristic biomarker profile in tissue and serum samples, exploitable for detecting and targeting accelerated aging in chronic hypoxic human diseases.
Collapse
Affiliation(s)
- Stefan Rudloff
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, A-1090 Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, A-1090 Vienna, Austria
| | - Nicola Wanner
- University Medical Center Hamburg-Eppendorf, III. Medizinische Klinik und Poliklinik, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Nastassia Liaukouskaya
- University Medical Center Hamburg-Eppendorf, III. Medizinische Klinik und Poliklinik, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Carsten Lundby
- Centre for Physical Activity Research (CFAS), Rigshospitalet Section 7641, Ole Maaloesvej 24, DK-2100 Copenhagen, Denmark; Faculty of Social and Health Sciences, Section for Health and Exercise Physiology, Inland Norway University of Applied Sciences, NO-2624 Lillehammer, Norway
| | - Tobias B Huber
- University Medical Center Hamburg-Eppendorf, III. Medizinische Klinik und Poliklinik, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, A-1090 Vienna, Austria.
| | - Uyen Huynh-Do
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland.
| |
Collapse
|
6
|
Rudloff S, Janot M, Rodriguez S, Dessalle K, Jahnen-Dechent W, Huynh-Do U. Fetuin-A is a HIF target that safeguards tissue integrity during hypoxic stress. Nat Commun 2021; 12:549. [PMID: 33483479 PMCID: PMC7822914 DOI: 10.1038/s41467-020-20832-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Intrauterine growth restriction (IUGR) is associated with reduced kidney size at birth, accelerated renal function decline, and increased risk for chronic kidney and cardiovascular diseases in adults. Precise mechanisms underlying fetal programming of adult diseases remain largely elusive and warrant extensive investigation. Setting up a mouse model of hypoxia-induced IUGR, fetal adaptations at mRNA, protein and cellular levels, and their long-term functional consequences are characterized, using the kidney as a readout. Here, we identify fetuin-A as an evolutionary conserved HIF target gene, and further investigate its role using fetuin-A KO animals and an adult model of ischemia-reperfusion injury. Beyond its role as systemic calcification inhibitor, fetuin-A emerges as a multifaceted protective factor that locally counteracts calcification, modulates macrophage polarization, and attenuates inflammation and fibrosis, thus preserving kidney function. Our study paves the way to therapeutic approaches mitigating mineral stress-induced inflammation and damage, principally applicable to all soft tissues.
Collapse
Affiliation(s)
- Stefan Rudloff
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Mathilde Janot
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Stephane Rodriguez
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Onco-haematology, Geneva Medical University, Geneva, Switzerland
| | - Kevin Dessalle
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Medical Faculty, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland.
| |
Collapse
|
7
|
Gobetto MN, Mendes Garrido Abregú F, Caniffi C, Veiras L, Elesgaray R, Gironacci M, Tomat AL, Arranz C. Fetal and postnatal zinc restriction: sex differences in the renal renin-angiotensin system of newborn and adult Wistar rats. J Nutr Biochem 2020; 81:108385. [PMID: 32388253 DOI: 10.1016/j.jnutbio.2020.108385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/05/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
This study aimed to evaluate renal morphology and the renal renin-angiotensin system in 6- and 81-day-old male and female offspring exposed to zinc deficiency during fetal life, lactation and/or postnatal growth. Female Wistar rats were fed low- or control zinc diets from pregnancy to offspring weaning. Afterwards, offspring were fed a low- or a control zinc diet until 81 days of life. In 6- and/or 81-day-old offspring, we evaluated systolic blood pressure, renal morphology, renal angiotensin II and angiotensin 1-7 concentration, and AT1 and AT2 receptors and angiotensin-converting enzymes protein and/or mRNA expression. At 6 days, zinc-deficient male offspring showed decreased glomerular filtration areas, remodelling of renal arteries, greater number of renal apoptotic cells, increased levels of Angiotensin II, higher Angiotensin II/Angiotensin 1-7 ratio and increased angiotensin-converting enzyme 1, AT1 and AT2 receptors mRNA and/or protein expression. Exacerbation of the renal Ang II/AT1 receptor axis and remodelling of renal arteries were also observed in adult zinc-deficient male offspring. An adequate zinc diet during post-weaning life did not improve all the alterations induced by zinc deficiency in early stages of development. Female offspring would appear to be less sensitive to zinc deficiency with no increase in blood pressure or significant alterations in renal morphology and the renin-angiotensin system. Moderate zinc deficiency during critical periods of prenatal and postnatal development leads to early morphological renal alterations and to permanent and long-term changes in the renal renin-angiotensin system that could predispose to renal and cardiovascular diseases in adult life.
Collapse
Affiliation(s)
- María Natalia Gobetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Facundo Mendes Garrido Abregú
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carolina Caniffi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Luciana Veiras
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Davis Research Bldg., Rm. 2007.110N, George Burns Rd., Los Angeles, CA 90048
| | - Rosana Elesgaray
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mariela Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Junín 956, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Junín 956, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| | - Analía Lorena Tomat
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Cristina Arranz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Fisiología, Junín 956, Piso 7, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina; CONICET, Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Junín 956, Piso 2, CP 1113, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
8
|
Cargill KR, Chiba T, Murali A, Mukherjee E, Crinzi E, Sims-Lucas S. Prenatal hypoxia increases susceptibility to kidney injury. PLoS One 2020; 15:e0229618. [PMID: 32084244 PMCID: PMC7034911 DOI: 10.1371/journal.pone.0229618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
Prenatal hypoxia is a gestational stressor that can result in developmental abnormalities or physiological reprogramming, and often decreases cellular capacity against secondary stress. When a developing fetus is exposed to hypoxia, blood flow is preferentially redirected to vital organs including the brain and heart over other organs including the kidney. Hypoxia-induced injury can lead to structural malformations in the kidney; however, even in the absence of structural lesions, hypoxia can physiologically reprogram the kidney leading to decreased function or increased susceptibility to injury. Our investigation in mice reveals that while prenatal hypoxia does not affect normal development of the kidneys, it primes the kidneys to have an increased susceptibility to kidney injury later in life. We found that our model does not develop structural abnormalities when prenatally exposed to modest 12% O2 as evident by normal histological characterization and gene expression analysis. Further, adult renal structure and function is comparable to mice exposed to ambient oxygen throughout nephrogenesis. However, after induction of kidney injury with a nephrotoxin (cisplatin), the offspring of mice housed in hypoxia exhibit significantly reduced renal function and proximal tubule damage following injury. We conclude that exposure to prenatal hypoxia in utero physiologically reprograms the kidneys leading to increased susceptibility to injury later in life.
Collapse
Affiliation(s)
- Kasey R. Cargill
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Takuto Chiba
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anjana Murali
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elina Mukherjee
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth Crinzi
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sunder Sims-Lucas
- Department of Pediatrics, Division of Nephrology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
9
|
Cargill K, Hemker SL, Clugston A, Murali A, Mukherjee E, Liu J, Bushnell D, Bodnar AJ, Saifudeen Z, Ho J, Bates CM, Kostka D, Goetzman ES, Sims-Lucas S. Von Hippel-Lindau Acts as a Metabolic Switch Controlling Nephron Progenitor Differentiation. J Am Soc Nephrol 2019; 30:1192-1205. [PMID: 31142573 PMCID: PMC6622426 DOI: 10.1681/asn.2018111170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Nephron progenitors, the cell population that give rise to the functional unit of the kidney, are metabolically active and self-renew under glycolytic conditions. A switch from glycolysis to mitochondrial respiration drives these cells toward differentiation, but the mechanisms that control this switch are poorly defined. Studies have demonstrated that kidney formation is highly dependent on oxygen concentration, which is largely regulated by von Hippel-Lindau (VHL; a protein component of a ubiquitin ligase complex) and hypoxia-inducible factors (a family of transcription factors activated by hypoxia). METHODS To explore VHL as a regulator defining nephron progenitor self-renewal versus differentiation, we bred Six2-TGCtg mice with VHLlox/lox mice to generate mice with a conditional deletion of VHL from Six2+ nephron progenitors. We used histologic, immunofluorescence, RNA sequencing, and metabolic assays to characterize kidneys from these mice and controls during development and up to postnatal day 21. RESULTS By embryonic day 15.5, kidneys of nephron progenitor cell-specific VHL knockout mice begin to exhibit reduced maturation of nephron progenitors. Compared with controls, VHL knockout kidneys are smaller and developmentally delayed by postnatal day 1, and have about half the number of glomeruli at postnatal day 21. VHL knockout nephron progenitors also exhibit persistent Six2 and Wt1 expression, as well as decreased mitochondrial respiration and prolonged reliance on glycolysis. CONCLUSIONS Our findings identify a novel role for VHL in mediating nephron progenitor differentiation through metabolic regulation, and suggest that VHL is required for normal kidney development.
Collapse
Affiliation(s)
- Kasey Cargill
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shelby L Hemker
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew Clugston
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Developmental Biology and
| | - Anjana Murali
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elina Mukherjee
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jiao Liu
- Section of Pediatric Nephrology, Department of Pediatrics and
- The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Daniel Bushnell
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew J Bodnar
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zubaida Saifudeen
- Section of Pediatric Nephrology, Department of Pediatrics and
- The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Jacqueline Ho
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carlton M Bates
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dennis Kostka
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Developmental Biology and
| | - Eric S Goetzman
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sunder Sims-Lucas
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania;
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Aiken CE, Tarry-Adkins JL, Spiroski AM, Nuzzo AM, Ashmore TJ, Rolfo A, Sutherland MJ, Camm EJ, Giussani DA, Ozanne SE. Chronic gestational hypoxia accelerates ovarian aging and lowers ovarian reserve in next-generation adult rats. FASEB J 2019; 33:7758-7766. [PMID: 30888848 PMCID: PMC6529349 DOI: 10.1096/fj.201802772r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Chronic fetal hypoxia is a common complication observed in human pregnancy, impacting pregnancies across global contexts. Exposure to chronic intrauterine hypoxia has major short- and long-term consequences for offspring health. However, the impact of chronic gestational hypoxia on female reproductive system development is unknown. We aimed to understand the impact of exposure to chronic fetal hypoxia on the developing female reproductive system. Wistar rat dams underwent normoxia (21%) or hypoxia (13%) during pregnancy. Postnatally, all female offspring were maintained in normoxic conditions into early adulthood. Female rats exposed to chronic gestational hypoxia (13%) during their intrauterine development had decreased ovarian primordial follicular reserve compared to controls (P < 0.05). Adult females who had been exposed to chronic fetal hypoxia had significantly reduced somatic ovarian telomere length (P < 0.05) and reduced ovarian protein expression of KU70, a critical component of the DNA-activated protein kinase repair complex (P < 0.01). Gene expression of NADPH oxidase 2-mediated oxidative stress markers was increased (P < 0.05). Exposure to chronic hypoxia during fetal development leads to accelerated aging of the somatic ovary and decreased ovarian reserve in adulthood. Ovarian aging is highly sensitive to gestational hypoxia, with implications for future fertility in next-generation offspring of high-risk pregnancies.-Aiken, C. E., Tarry-Adkins, J. L., Spiroski, A.-M., Nuzzo, A. M., Ashmore, T. J., Rolfo, A., Sutherland, M. J., Camm, E. J., Giussani, D. A., Ozanne, S. E. Chronic gestational hypoxia accelerates ovarian aging and lowers ovarian reserve in next-generation adult rats.
Collapse
Affiliation(s)
- Catherine E. Aiken
- University of Cambridge Metabolic Research Laboratories and Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome Trust–MRC Institute of Metabolic Science, Addenbrooke’s Treatment Centre, Addenbrooke’s Hospital, Cambridge, United Kingdom;,Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, United Kingdom;,Correspondence: University of Cambridge, Hills Rd, Cambridge CB2 0QQ, United Kingdom. E-mail:
| | - Jane L. Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome Trust–MRC Institute of Metabolic Science, Addenbrooke’s Treatment Centre, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Ana-Mishel Spiroski
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Anna M. Nuzzo
- Dipartimento di Scienze Chirurgiche, Universita degli Studi di Torino, Turin, Italy
| | - Thomas J. Ashmore
- University of Cambridge Metabolic Research Laboratories and Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome Trust–MRC Institute of Metabolic Science, Addenbrooke’s Treatment Centre, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Alessandro Rolfo
- Dipartimento di Scienze Chirurgiche, Universita degli Studi di Torino, Turin, Italy
| | - Megan J. Sutherland
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Emily J. Camm
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Dino A. Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome Trust–MRC Institute of Metabolic Science, Addenbrooke’s Treatment Centre, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
11
|
Vieira-Rocha M, Rodríguez-Rodríguez P, Sousa J, González M, Arribas S, López de Pablo A, Diniz C. Vascular angiotensin AT1 receptor neuromodulation in fetal programming of hypertension. Vascul Pharmacol 2019; 117:27-34. [DOI: 10.1016/j.vph.2018.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/31/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022]
|
12
|
Aiken CE, Tarry‐Adkins JL, Spiroski A, Nuzzo AM, Ashmore TJ, Rolfo A, Sutherland MJ, Camm EJ, Giussani DA, Ozanne SE. Chronic fetal hypoxia disrupts the peri-conceptual environment in next-generation adult female rats. J Physiol 2019; 597:2391-2401. [PMID: 30791124 PMCID: PMC6487938 DOI: 10.1113/jp277431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/15/2019] [Indexed: 02/02/2023] Open
Abstract
KEY POINTS Exposure to chronic hypoxia during gestation influences long-term health and development, including reproductive capacity, across generations. If the peri-conceptual environment in the developing oviduct is affected by gestational hypoxia, then this could have implications for later fertility and the health of future generations. In the present study, we show that the oviducts of female rats exposed to chronic hypoxia in utero have reduced telomere length, decreased mitochondrial DNA biogenesis and increased oxidative stress The results of the present study show that exposure to chronic gestational hypoxia leads to accelerated ageing of the oviduct in early adulthood and they help us understand how exposure to hypoxia during development could influence reproductive health across generations. ABSTRACT Exposure to chronic hypoxia during fetal development has important effects on immediate and long-term outcomes in offspring. Adverse impacts in adult offspring include impairment of cardiovascular function, metabolic derangement and accelerated ovarian ageing. However, it is not known whether other aspects of the female reproductive system may be similarly affected. In the present study, we examined the impact of chronic gestational hypoxia on the developing oviduct. Wistar rat dams were randomized to either normoxia (21%) or hypoxia (13%) from day 6 post-mating until delivery. Post-delivery female offspring were maintained in normoxia until 4 months of age. Oviductal gene expression was assayed at the RNA (quantitative RT-PCR) and protein (western blotting) levels. Oviductal telomere length was assayed using Southern blotting. Oviductal telomere length was reduced in the gestational hypoxia-exposed animals compared to normoxic controls (P < 0.01). This was associated with a specific post-transcriptional reduction in the KU70 subunit of DNA-pk in the gestational hypoxia-exposed group (P < 0.05). Gestational hypoxia-exposed oviducts also showed evidence of decreased mitochondrial DNA biogenesis, reduced mtDNA copy number (P < 0.05) and reduced gene expression of Tfam (P < 0.05) and Pgc1α (P < 0.05). In the hypoxia-exposed oviducts, there was upregulation of mitochondrial-specific anti-oxidant defence enzymes (MnSOD; P < 0.01). Exposure to chronic gestational hypoxia leads to accelerated ageing of the oviduct in adulthood. The oviduct plays a central role in early development as the site of gamete transport, syngamy, and early development; hence, accelerated ageing of the oviductal environment could have important implications for fertility and the health of future generations.
Collapse
Affiliation(s)
- Catherine E. Aiken
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases UnitWellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's HospitalCambridgeUK
- University Department of Obstetrics and GynaecologyUniversity of Cambridge, CambridgeUK
| | - Jane L. Tarry‐Adkins
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases UnitWellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's HospitalCambridgeUK
| | - Ana‐Mishel Spiroski
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Anna M. Nuzzo
- Dipartimento di Scienze ChirurgicheUniversita degli Studi di TorinoTurinItaly
| | - Thomas J. Ashmore
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases UnitWellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's HospitalCambridgeUK
| | - Alessandro Rolfo
- Dipartimento di Scienze ChirurgicheUniversita degli Studi di TorinoTurinItaly
| | - Megan J. Sutherland
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Emily J. Camm
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Dino A. Giussani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases UnitWellcome Trust‐MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's HospitalCambridgeUK
| |
Collapse
|
13
|
Zhong Y, Feng X, Xu T, Yang C, Zhang W, Chen X, Fan X, Lu L, Zhang M, Li L, Xu Z. Inherited risk plus prenatal insult caused malignant dysfunction in mesenteric arteries in adolescent SHR offspring. PLoS One 2019; 14:e0215994. [PMID: 31017969 PMCID: PMC6481862 DOI: 10.1371/journal.pone.0215994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/11/2019] [Indexed: 11/19/2022] Open
Abstract
Prenatal hypoxia can induce cardiovascular diseases in the offspring. This study determined whether and how prenatal hypoxia may cause malignant hypertension and impaired vascular functions in spontaneous hypertension rat (SHR) offspring at adolescent stage. Pregnant SHR were placed in a hypoxic chamber (11% O2) or normal environment (21% O2) from gestational day 6 until birth. Body weight and blood pressure (BP) of SHR offspring were measured every week from 5 weeks old. Mesenteric arteries were tested. Gestational hypoxia resulted in growth restriction during 6-12 weeks and a significant elevation in systolic pressure in adolescent offspring at 12 weeks old. Notably, endothelial vasodilatation of mesenteric arteries was impaired in SHR adolescent offspring exposed to prenatal hypoxia, vascular responses to acetylcholine (ACh) and sodium nitroprusside (SNP) were reduced, as well as plasma nitric oxide levels and expression of endothelial nitric oxide synthase (eNOS) in vessels were decreased. Moreover, mesenteric arteries in SHR offspring following prenatal hypoxia showed enhanced constriction responses to phenylephrine (PE), associated with up-regulated activities of L-type calcium channel (Ca2+-dependent), RhoA/Rock pathway signaling (Ca2+-sensitization), and intracellular Ca2+ flow. Pressurized myograph demonstrated altered mechanical properties with aggravated stiffness in vessels, while histological analysis revealed vascular structural disorganization in prenatal hypoxia offspring. The results demonstrated that blood pressure and vascular function in young SHR offspring were affected by prenatal hypoxia, providing new information on development of hypertension in adolescent offspring with inherited hypertensive backgrounds.
Collapse
Affiliation(s)
- Yuan Zhong
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
- Obstetrics and Gynecology, Municipal Hospital, Suzhou, Jiangsu, China
| | - Xueqin Feng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ting Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chunli Yang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wenna Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xueyi Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaorong Fan
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Likui Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Meng Zhang
- Obstetrics and Gynecology, Tengzhou Central People’s Hospital, Zaozhuang, Shandong, China
| | - Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (ZX); (LL)
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, Jiangsu, China
- Center for Prenatal Biology, Loma Linda University, Loma Linda, CA, United States of America
- * E-mail: (ZX); (LL)
| |
Collapse
|
14
|
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
|
15
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
16
|
Zhu Y, Zuo N, Li B, Xiong Y, Chen H, He H, Sun Z, Hu S, Cheng H, Ao Y, Wang H. The expressional disorder of the renal RAS mediates nephrotic syndrome of male rat offspring induced by prenatal ethanol exposure. Toxicology 2018; 400-401:9-19. [PMID: 29548890 DOI: 10.1016/j.tox.2018.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/03/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022]
Abstract
This study aimed to prove that prenatal ethanol exposure (PEE) can induce nephrotic syndrome in male rat offspring and to explore the underlying intrauterine programming mechanisms. Pregnant Wistar rats were intragastrically administered ethanol (4 g/kg d) from gestational day (GD) 9 to GD 20, and the male fetuses were delivered by cesarean section at GD20 and the male adult offspring were euthanized at postnatal week (PW) 24. In vitro, the primary metanephric mesenchyme cells were treated with ethanol at concentrations of 15-60 mM. The results indicated that the kidneys of adult offspring in the PEE group exhibited glomerulosclerosis as well as interstitial fibrosis. The levels of serum creatinine and urine protein were elevated; the serum total cholesterol level was increased and the serum albumin concentration was reduced. In the fetal kidney, developmental retardation was presented in the PEE group via pathological examinations, accompanied by the expressional inhibition of the glial-cell-line-derived neurotrophic factor/c-ret tyrosine kinase receptor (GDNF/c-ret) signaling pathway. Although serum angiotensin II (Ang II) level and the gene expression of renal angiotensin-converting enzyme (ACE) were increased in the PEE group, the expression of renal angiotensin II type 2 receptor (AT2R) was significantly inhibited, accompanied by a reduction in the H3K27ac level on the AT2R gene promoter. In the non-classical renin-angiotensin system (RAS), the expression of renal angiotensin converting enzyme 2 (ACE2) and Mas receptor (MasR) were inhibited in the PEE group. The above changes of the classical and non-classical RAS all sustained from utero to adulthood. In vitro, ethanol elevated the gene expression of ACE and angiotensin II type 1a receptor (AT1aR) whereas it reduced the expression of AT2R, ACE2, and MasR, accompanied by a reduction in the H3K27ac level on AT2R gene promoter. Taken together, these results suggested that PEE can induce fetal kidney developmental retardation and adult nephrotic syndrome, and direct regulation of ethanol to the renal RAS was involved in the mechanism of nephrotic syndrome induced by PEE.
Collapse
Affiliation(s)
- Yanan Zhu
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Na Zuo
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Ying Xiong
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Haiyun Chen
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Hangyuan He
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Zhaoxia Sun
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Shuangshuang Hu
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China
| | - Hui Cheng
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ying Ao
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Science of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan, 430071, China.
| |
Collapse
|
17
|
Kobayashi H, Liu J, Urrutia AA, Burmakin M, Ishii K, Rajan M, Davidoff O, Saifudeen Z, Haase VH. Hypoxia-inducible factor prolyl-4-hydroxylation in FOXD1 lineage cells is essential for normal kidney development. Kidney Int 2017; 92:1370-1383. [PMID: 28847650 PMCID: PMC5696043 DOI: 10.1016/j.kint.2017.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 05/26/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022]
Abstract
Hypoxia in the embryo is a frequent cause of intra-uterine growth retardation, low birth weight, and multiple organ defects. In the kidney, this can lead to low nephron endowment, predisposing to chronic kidney disease and arterial hypertension. A key component in cellular adaptation to hypoxia is the hypoxia-inducible factor pathway, which is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3. In the adult kidney, PHD oxygen sensors are differentially expressed in a cell type-dependent manner and control the production of erythropoietin in interstitial cells. However, the role of interstitial cell PHDs in renal development has not been examined. Here we used a genetic approach in mice to interrogate PHD function in FOXD1-expressing stroma during nephrogenesis. We demonstrate that PHD2 and PHD3 are essential for normal kidney development as the combined inactivation of stromal PHD2 and PHD3 resulted in renal failure that was associated with reduced kidney size, decreased numbers of glomeruli, and abnormal postnatal nephron formation. In contrast, nephrogenesis was normal in animals with individual PHD inactivation. We furthermore demonstrate that the defect in nephron formation in PHD2/PHD3 double mutants required intact hypoxia-inducible factor-2 signaling and was dependent on the extent of stromal hypoxia-inducible factor activation. Thus, hypoxia-inducible factor prolyl-4-hydroxylation in renal interstitial cells is critical for normal nephron formation.
Collapse
Affiliation(s)
- Hanako Kobayashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Jiao Liu
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, Louisiana, USA; The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Andres A Urrutia
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mikhail Burmakin
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Ken Ishii
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Malini Rajan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Olena Davidoff
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Zubaida Saifudeen
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, Louisiana, USA; The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Volker H Haase
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA; Department of Cancer Biology and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
18
|
Dorri Mashhadi F, Zavvar Reza J, Jamhiri M, Hafizi Z, Zare Mehrjardi F, Safari F. The effect of resveratrol on angiotensin II levels and the rate of transcription of its receptors in the rat cardiac hypertrophy model. J Physiol Sci 2017; 67:303-309. [PMID: 27324786 PMCID: PMC10717376 DOI: 10.1007/s12576-016-0465-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/03/2016] [Indexed: 10/21/2022]
Abstract
This study investigated the effect of resveratrol on serum and cardiac levels of angiotensin II and transcription of its main receptors following pressure overload induced-hypertrophy. Rats were divided into untreated (Hyp) and resveratrol treated hypertrophied groups (H + R). Intact animals served as the control (Ctl). Cardiac hypertrophy was induced by abdominal aortic banding. Blood pressure (BP) was recorded via left carotid artery cannula. Fibrosis was confirmed by Masson trichrome staining. Angiotensin II level was measured using an ELIZA test. Gene expression was assessed by a real time PCR (RT-PCR) technique. We observed that in the H + R group BP and heart weight/body weight were decreased significantly (p < 0.001, p < 0.05, respectively vs Hyp). The cardiac levels of angiotensin II and AT1a mRNA were increased in the Hyp group (p < 0.01 vs Ctl). In the H + R group the AT1a mRNA level was decreased significantly (p < 0.05 vs Hyp). It could be concluded that resveratrol protects the heart against hypertrophy progression in part by affecting cardiac AT1a transcription.
Collapse
Affiliation(s)
- Fahimeh Dorri Mashhadi
- Department of Medical Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Zavvar Reza
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohabbat Jamhiri
- Department of Medical Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeinab Hafizi
- Department of Medical Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Zare Mehrjardi
- Department of Medical Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Medical Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Kidney development depends on outgrowth of the ureteric bud into the metanephric mesenchyme. The number of ureteric bud branching events determines the final number of nephrons, which correlates inversely with the risk for development of chronic kidney disease and arterial hypertension during lifetime. The purpose of this review is to highlight the influence of oxygen on nephrogenesis and to describe cellular mechanisms by which hypoxia can impair nephron formation. RECENT FINDINGS Although kidney development normally takes place under hypoxic conditions, nephrogenesis is impaired when oxygen availability falls below the usual range. Hypoxia-inducible factors (HIF) play an important role in linking low oxygen concentrations to the biology of nephron formation, but their effect appears to be cell type dependent. In ureteric bud cells, HIF stimulates tubulogenesis, whereas HIF stabilization in cells of the metanephric mesenchyme results in secretion of growth factors, including vascular endothelial growth factor A, which in aggregate inhibit ureteric bud branching. The balance between pro and antibranching effects may be altered in various ways, but the inhibitory effect usually seems to predominate under reduced oxygen concentrations, explaining how intrauterine hypoxia can lead to low nephron numbers. SUMMARY Oxygen availability has a complex influence on nephrogenesis. Oxygen concentrations outside an optimal low range may affect nephron endowment. Associations between placental insufficiency and increased risk for chronic kidney disease and arterial hypertension during later life may to a large extent be due to direct effects of reduced oxygen supply to the metanephric mesenchyme and mediated through the HIF pathway.
Collapse
|
20
|
Effect of Hypoxia on the Differentiation and the Self-Renewal of Metanephrogenic Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:7168687. [PMID: 28194187 PMCID: PMC5282446 DOI: 10.1155/2017/7168687] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/24/2016] [Accepted: 12/07/2016] [Indexed: 12/31/2022] Open
Abstract
Hypoxia is an important and influential factor in development. The embryonic kidney is exposed to a hypoxic environment throughout its development. The Wnt/β-catenin pathway plays vital roles in the differentiation and self-renewal of metanephrogenic mesenchymal stem cells (MMSCs) from which the kidney is derived. Thus, we hypothesized that hypoxia can regulate the differentiation and pluripotency of MMSCs through the Wnt/β-catenin pathway. To test this hypothesis, MMSCs from rats at embryonic day 18.5 were cultured in normoxic (21% O2) and hypoxic (1% O2) conditions. The effects of hypoxia on differentiation, stemness, proliferation, and apoptosis of cultured MMSCs and on the activity of the Wnt/β-catenin pathway were tested. Our results revealed that the hypoxic condition increased the number of epithelial cells (E-cadherin+ or CK18+) as well the expression of markers of renal tubule epithelia cells (CDH6, Aqp1, and OPN), decreased the number and proliferation of stem cells (SIX-2+ or CITED1+), and induced apoptosis. Additionally, hypoxia reduced the expression of Wnt4 as well as its downstream molecules β-catenin, LEF-1, and Axin2. Activation of the Wnt/β-catenin pathway by LiCl or BIO modified the effects of hypoxia on the differentiation and self-renewal of MMSCs. Thus, we concluded that hypoxia induces the differentiation and inhibits the self-renewal of MMSCs by inhibiting the Wnt/β-catenin pathway. The observations further our understanding of the effects of hypoxia on kidney.
Collapse
|
21
|
Kranch-Shorthouse RA, Bauer AS, Magness RR, Lopez GE, Segar JL, Blohowiak SE, Kling PJ. Ovine uterine space restriction causes dysregulation of the renin-angiotensin system in fetal kidneys. Biol Reprod 2017; 96:211-220. [PMID: 28395333 DOI: 10.1095/biolreprod.116.140079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 12/06/2016] [Indexed: 11/01/2022] Open
Abstract
In ovine pregnancy, uterine space restriction (USR) resulting from decreased space for placental attachment caused intrauterine growth restriction and impaired nephrogenesis. The fetal kidney renin-angiotensin system (RAS) is involved in nephrogenesis, fluid balance, and iron deposition. Angiotensin II exerts its effects via multiple receptors: angiotensin II 1-8 receptor type 1 (AT 1 R) and type 2 (AT 2 R), and angiotensin II 1-7 Mas receptor (MASR). Objective : To test the hypothesis that ovine USR is associated with dysregulation of the fetal renal RAS. Methods : Multiparous pregnant ewes (n = 32), 16 with surgical bifurcated disconnection of one uterine horn to further reduce placental attachment sites, were studied. USR (n = 31) ovine fetuses were compared to nonspace restricted (NSR) singleton controls (n = 22) on gestational day (GD) 120 or GD130, term GD147. Fetal plasma was collected to evaluate plasma renin activity and iron indices. Fetal kidney AT 1 R, AT 2 R, and MASR proteins were assessed by Western immunoblotting and immunohistochemistry. Results : AT 1 R, AT 2 R, and MASR protein expression was higher in USR at GD130 than aged-matched NSR and USR at GD120, ( P < 0.05 all). AT 1 R and AT 2 R localization was homogenous throughout proximal and distal tubules in both USR and NSR at both gestational dates. MASR localization was punctate throughout renal cortical structures including tubules and glomeruli in both USR and NSR, shifted to intranuclear at GD130. Plasma renin activity was inversely related to plasma osmolarity ( P < 0.02) and was downregulated in USR at GD130 ( P < 0.05). Conclusions : By late gestation, USR upregulated renal angiotensin receptor expression, an effect with potential functional implications.
Collapse
Affiliation(s)
- Rachel A Kranch-Shorthouse
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin, USA.,Department of Nutritional Science, The University of Arizona, Arizona, USA
| | - Adam S Bauer
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin, USA
| | - Ronald R Magness
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Wisconsin, USA.,Department of Animal Sciences, University of Wisconsin-Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, University of South Florida, Florida, USA
| | - Gladys E Lopez
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Wisconsin, USA
| | | | - Sharon E Blohowiak
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin, USA
| | - Pamela J Kling
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
22
|
Zhang X, Zhou X, Li L, Sun M, Gao Q, Zhang P, Tang J, He Y, Zhu D, Xu Z. Chronic hypoxia in pregnancy affects thymus development in Balb/c mouse offspring via IL2 Signaling. Mol Reprod Dev 2016; 83:337-46. [PMID: 26918321 DOI: 10.1002/mrd.22630] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/29/2016] [Indexed: 01/15/2023]
Affiliation(s)
- Xiaopeng Zhang
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Xiuwen Zhou
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Lingjun Li
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Miao Sun
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Qingqing Gao
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Pengjie Zhang
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Jiaqi Tang
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Yu He
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Di Zhu
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
| | - Zhice Xu
- Institute for Fetology; First Hospital of Soochow University; Suzhou China
- Center for Perinatal Biology; Loma Linda University; California
| |
Collapse
|
23
|
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
|
24
|
Chang EI, Zárate MA, Rabaglino MB, Richards EM, Keller-Wood M, Wood CE. Ketamine suppresses hypoxia-induced inflammatory responses in the late-gestation ovine fetal kidney cortex. J Physiol 2015; 594:1295-310. [PMID: 26497972 DOI: 10.1113/jp271066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/28/2015] [Indexed: 01/04/2023] Open
Abstract
Acute fetal hypoxia is a form of fetal stress that stimulates renal vasoconstriction and ischaemia as a consequence of the physiological redistribution of combined ventricular output. Because of the potential ischaemia-reperfusion injury to the kidney, we hypothesized that it would respond to hypoxia with an increase in the expression of inflammatory genes, and that ketamine (an N-methyl-D-aspartate receptor antagonist) would reduce or block this response. Hypoxia was induced for 30 min in chronically catheterized fetal sheep (125 ± 3 days), with or without ketamine (3 mg kg(-1)) administered intravenously to the fetus 10 min prior to hypoxia. Gene expression in fetal kidney cortex collected 24 h after the onset of hypoxia was analysed using ovine Agilent 15.5k array and validated with qPCR and immunohistochemistry in four groups of ewes: normoxic control, normoxia + ketamine, hypoxic control and hypoxia + ketamine (n = 3-4 per group). Significant differences in gene expression between groups were determined with t-statistics using the limma package for R (P ≤ 0.05). Enriched biological processes for the 427 upregulated genes were immune and inflammatory responses and for the 946 downregulated genes were metabolic processes. Ketamine countered the effects of hypoxia on upregulated immune/inflammatory responses as well as the downregulated metabolic responses. We conclude that our transcriptomics modelling predicts that hypoxia activates inflammatory pathways and reduces metabolism in the fetal kidney cortex, and ketamine blocks or ameliorates this response. The results suggest that ketamine may have therapeutic potential for protection from ischaemic renal damage.
Collapse
Affiliation(s)
- Eileen I Chang
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610-0274, USA
| | - Miguel A Zárate
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610-0274, USA
| | - Maria B Rabaglino
- Department of Animal Reproduction, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Elaine M Richards
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, 32610-0487, USA
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, 32610-0487, USA
| | - Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610-0274, USA
| |
Collapse
|
25
|
Chronic hypoxia in pregnancy affected vascular tone of renal interlobar arteries in the offspring. Sci Rep 2015; 5:9723. [PMID: 25983078 PMCID: PMC4434890 DOI: 10.1038/srep09723] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/17/2015] [Indexed: 11/22/2022] Open
Abstract
Hypoxia during pregnancy could affect development of fetuses as well as cardiovascular systems in the offspring. This study was the first to demonstrate the influence and related mechanisms of prenatal hypoxia (PH) on renal interlobar arteries (RIA) in the 5-month-old male rat offspring. Following chronic hypoxia during pregnancy, phenylephrine induced significantly higher pressor responses and greater vasoconstrictions in the offspring. Nitric oxide mediated vessel relaxation was altered in the RIA. Phenylephrine-stimulated free intracellular calcium was significantly higher in the RIA of the PH group. The activity and expression of L-type calcium channel (Cav1.2), not T-type calcium channel (Cav3.2), was up-regulated. The whole-cell currents of calcium channels and the currents of Cav1.2 were increased compared with the control. In addition, the whole-cell K+ currents were decreased in the offspring exposed to prenatal hypoxia. Activity of large-conductance Ca2+-activated K+ channels and the expression of MaxiKα was decreased in the PH group. The results provide new information regarding the influence of prenatal hypoxia on the development of the renal vascular system, and possible underlying cellular and ion channel mechanisms involved.
Collapse
|
26
|
Xia S, Lv J, Gao Q, Li L, Chen N, Wei X, Xiao J, Chen J, Tao J, Sun M, Mao C, Zhang L, Xu Z. Prenatal exposure to hypoxia induced Beclin 1 signaling-mediated renal autophagy and altered renal development in rat fetuses. Reprod Sci 2014; 22:156-64. [PMID: 24872334 DOI: 10.1177/1933719114536474] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
AIMS Hypoxia has adverse effects on renal development. This study was the first to test hypoxia-induced renal autophagy in rat fetuses. METHODS Pregnant rats were exposed to hypoxia or normoxia during pregnancy and fetal kidneys were collected at gestation day 21. RESULTS Fetal kidney weight and ratio of kidney-body weight were reduced. Histological analysis showed enlargement in Bowman space and wider space between interstitia in the kidneys of fetus exposed to hypoxia. Fetal renal B-cell lymphoma 2 (BCL-2) was decreased accompanied with higher 2'-deoxyuridine 5'-triphosphate nick end-labeling staining and unchanged soluble FAS in the hypoxia group. Hypoxia increased autophagic structures, including autophagosomes and autolysosomes, in fetal kidneys and increased renal APG5L. There was an increase in renal LC3-II, Beclin 1, p-S6, hypoxia inducible factor 1α (HIF-1a), and ratio of LC3-II-LC3-I and a decrease in P62, protein kinase B (AKT), and phosphorylated AKT in the hypoxia group. Both renal mammalian target of rapamycin (mTOR) and Beclin 1 signaling were upregulated. CONCLUSION Hypoxia-affected fetal renal development was associated with renal apoptosis and Beclin 1 signaling-mediated autophagy.
Collapse
Affiliation(s)
- Shuixiu Xia
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Juanxiu Lv
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Ningjing Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiaoguang Wei
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jianping Xiao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jie Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jianying Tao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lubo Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
27
|
Cuffe JSM, Walton SL, Singh RR, Spiers JG, Bielefeldt-Ohmann H, Wilkinson L, Little MH, Moritz KM. Mid- to late term hypoxia in the mouse alters placental morphology, glucocorticoid regulatory pathways and nutrient transporters in a sex-specific manner. J Physiol 2014; 592:3127-41. [PMID: 24801305 DOI: 10.1113/jphysiol.2014.272856] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Maternal hypoxia is a common perturbation that can disrupt placental and thus fetal development, contributing to neonatal impairments. Recently, evidence has suggested that physiological outcomes are dependent upon the sex of the fetus, with males more susceptible to hypoxic insults than females. This study investigated the effects of maternal hypoxia during mid- to late gestation on fetal growth and placental development and determined if responses were sex specific. CD1 mice were housed under 21% or 12% oxygen from embryonic day (E) 14.5 until tissue collection at E18.5. Fetuses and placentas were weighed before collection for gene and protein expression and morphological analysis. Hypoxia reduced fetal weight in both sexes at E18.5 by 7% but did not affect placental weight. Hypoxia reduced placental mRNA levels of the mineralocorticoid and glucocorticoid receptors and reduced the gene and protein expression of the glucocorticoid metabolizing enzyme HSD11B2. However, placentas of female fetuses responded differently to maternal hypoxia than did placentas of male fetuses. Notably, morphology was significantly altered in placentas from hypoxic female fetuses, with a reduction in placental labyrinth blood spaces. In addition mRNA expression of Glut1, Igf2 and Igf1r were reduced in placentas of female fetuses only. In summary, maternal hypoxia altered placental formation in a sex specific manner through mechanisms involving placental vascular development, growth factor and nutrient transporter expression and placental glucocorticoid signalling. This study provides insight into how sex differences in offspring disease development may be due to sex specific placental adaptations to maternal insults.
Collapse
Affiliation(s)
- J S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - S L Walton
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - R R Singh
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - J G Spiers
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - H Bielefeldt-Ohmann
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - L Wilkinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - M H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|