1
|
Cortes-Araya Y, Cheung S, Ho W, Stenhouse C, Ashworth CJ, Esteves CL, Donadeu FX. Effects of foetal size, sex and developmental stage on adaptive transcriptional responses of skeletal muscle to intrauterine growth restriction in pigs. Sci Rep 2024; 14:8500. [PMID: 38605102 PMCID: PMC11009347 DOI: 10.1038/s41598-024-57194-9] [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: 06/23/2023] [Accepted: 03/15/2024] [Indexed: 04/13/2024] Open
Abstract
Intrauterine growth restriction (IUGR) occurs both in humans and domestic species. It has a particularly high incidence in pigs, and is a leading cause of neonatal morbidity and mortality as well as impaired postnatal growth. A key feature of IUGR is impaired muscle development, resulting in decreased meat quality. Understanding the developmental origins of IUGR, particularly at the molecular level, is important for developing effective strategies to mitigate its economic impact on the pig industry and animal welfare. The aim of this study was to characterise transcriptional profiles in the muscle of growth restricted pig foetuses at different gestational days (GD; gestational length ~ 115 days), focusing on selected genes (related to development, tissue injury and metabolism) that were previously identified as dysregulated in muscle of GD90 fetuses. Muscle samples were collected from the lightest foetus (L) and the sex-matched foetus with weight closest to the litter average (AW) from each of 22 Landrace x Large White litters corresponding to GD45 (n = 6), GD60 (n = 8) or GD90 (n = 8), followed by analyses, using RT-PCR and protein immunohistochemistry, of selected gene targets. Expression of the developmental genes, MYOD, RET and ACTN3 were markedly lower, whereas MSTN expression was higher, in the muscle of L relative to AW littermates beginning on GD45. Levels of all tissue injury-associated transcripts analysed (F5, PLG, KNG1, SELL, CCL16) were increased in L muscle on GD60 and, most prominently, on GD90. Among genes involved in metabolic regulation, KLB was expressed at higher levels in L than AW littermates beginning on GD60, whereas both IGFBP1 and AHSG were higher in L littermates on GD90 but only in males. Furthermore, the expression of genes specifically involved in lipid, hexose sugar or iron metabolism increased or, in the case of UCP3, decreased in L littermates on GD60 (UCP3, APOB, ALDOB) or GD90 (PNPLA3, TF), albeit in the case of ALDOB this only involved females. In conclusion, marked dysregulation of genes with critical roles in development in L foetuses can be observed from GD45, whereas for a majority of transcripts associated with tissue injury and metabolism differences between L and AW foetuses were apparent by GD60 or only at GD90, thus identifying different developmental windows for different types of adaptive responses to IUGR in the muscle of porcine foetuses.
Collapse
Affiliation(s)
- Y Cortes-Araya
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - S Cheung
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - W Ho
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - C Stenhouse
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
- Department of Animal Science, Pennsylvania State University, State College, PA, 16803, USA
| | - C J Ashworth
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - C L Esteves
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - F X Donadeu
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK.
| |
Collapse
|
2
|
Voggel J, Fink G, Zelck M, Wohlfarth M, Post JM, Bindila L, Rauh M, Amann K, Alejandre Alcázar MA, Dötsch J, Nüsken KD, Nüsken E. Elevated n-3/n-6 PUFA ratio in early life diet reverses adverse intrauterine kidney programming in female rats. J Lipid Res 2022; 63:100283. [PMID: 36152882 PMCID: PMC9619183 DOI: 10.1016/j.jlr.2022.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/11/2022] [Indexed: 11/27/2022] Open
Abstract
Intrauterine growth restriction (IUGR) predisposes to chronic kidney disease via activation of proinflammatory pathways, and omega-3 PUFAs (n-3 PUFAs) have anti-inflammatory properties. In female rats, we investigated 1) how an elevated dietary n-3/n-6 PUFA ratio (1:1) during postnatal kidney development modifies kidney phospholipid (PL) and arachidonic acid (AA) metabolite content and 2) whether the diet counteracts adverse molecular protein signatures expected in IUGR kidneys. IUGR was induced by bilateral uterine vessel ligation or intrauterine stress through sham operation 3.5 days before term. Control (C) offspring were born after uncompromised pregnancy. On postnatal (P) days P2–P39, rats were fed control (n-3/n-6 PUFA ratio 1:20) or n-3 PUFA intervention diet (N3PUFA; ratio 1:1). Plasma parameters (P33), kidney cortex lipidomics and proteomics, as well as histology (P39) were studied. We found that the intervention diet tripled PL-DHA content (PC 40:6; P < 0.01) and lowered both PL-AA content (PC 38:4 and lyso-phosphatidylcholine 20:4; P < 0.05) and AA metabolites (HETEs, dihydroxyeicosatrienoic acids, and epoxyeicosatrienoic acids) to 25% in all offspring groups. After ligation, our network analysis of differentially expressed proteins identified an adverse molecular signature indicating inflammation and hypercoagulability. N3PUFA diet reversed 61 protein alterations (P < 0.05), thus mitigating adverse IUGR signatures. In conclusion, an elevated n-3/n-6 PUFA ratio in early diet strongly reduces proinflammatory PLs and mediators while increasing DHA-containing PLs regardless of prior intrauterine conditions. Counteracting a proinflammatory hypercoagulable protein signature in young adult IUGR individuals through early diet intervention may be a feasible strategy to prevent developmentally programmed kidney damage in later life.
Collapse
Affiliation(s)
- Jenny Voggel
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gregor Fink
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Magdalena Zelck
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Maria Wohlfarth
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Julia M Post
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Miguel A Alejandre Alcázar
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Jörg Dötsch
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Kai-Dietrich Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Eva Nüsken
- Clinic and Polyclinic for Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany.
| |
Collapse
|
3
|
Intrauterine growth restriction: Clinical consequences on health and disease at adulthood. Reprod Toxicol 2021; 99:168-176. [DOI: 10.1016/j.reprotox.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
|
4
|
Parra-Vargas M, Ramon-Krauel M, Lerin C, Jimenez-Chillaron JC. Size Does Matter: Litter Size Strongly Determines Adult Metabolism in Rodents. Cell Metab 2020; 32:334-340. [PMID: 32814016 DOI: 10.1016/j.cmet.2020.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/16/2020] [Accepted: 07/20/2020] [Indexed: 11/25/2022]
Abstract
In this essay, we highlight how litter size in rodents is a strong determinant of neonatal growth and long-term metabolic health. Based on these effects, we strongly advise that scientific articles that utilize rodent models for obesity and metabolic research should include information on the litter sizes in the study to increase the data transparency of such reports.
Collapse
Affiliation(s)
- Marcela Parra-Vargas
- Institut de Recerca Sant Joan de Déu, Endocrinology, c/ Santa Rosa 39-57, 4ª planta, Esplugues, 08950 Barcelona, Spain
| | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu, Endocrinology, c/ Santa Rosa 39-57, 4ª planta, Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu, Endocrinology, c/ Santa Rosa 39-57, 4ª planta, Esplugues, 08950 Barcelona, Spain
| | - Josep C Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu, Endocrinology, c/ Santa Rosa 39-57, 4ª planta, Esplugues, 08950 Barcelona, Spain.
| |
Collapse
|
5
|
Torreggiani M, Fois A, D’Alessandro C, Colucci M, Orozco Guillén AO, Cupisti A, Piccoli GB. Of Mice and Men: The Effect of Maternal Protein Restriction on Offspring's Kidney Health. Are Studies on Rodents Applicable to Chronic Kidney Disease Patients? A Narrative Review. Nutrients 2020; 12:E1614. [PMID: 32486266 PMCID: PMC7352514 DOI: 10.3390/nu12061614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/19/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
In the almost 30 years that have passed since the postulation of the "Developmental Origins of Health and Disease" theory, it has been clearly demonstrated that a mother's dietary habits during pregnancy have potential consequences for her offspring that go far beyond in utero development. Protein malnutrition during pregnancy, for instance, can cause severe alterations ranging from intrauterine growth retardation to organ damage and increased susceptibility to hypertension, diabetes mellitus, cardiovascular diseases and chronic kidney disease (CKD) later in life both in experimental animals and humans. Conversely, a balanced mild protein restriction in patients affected by CKD has been shown to mitigate the biochemical derangements associated with kidney disease and even slow its progression. The first reports on the management of pregnant CKD women with a moderately protein-restricted plant-based diet appeared in the literature a few years ago. Today, this approach is still being debated, as is the optimal source of protein during gestation in CKD. The aim of this report is to critically review the available literature on the topic, focusing on the similarities and differences between animal and clinical studies.
Collapse
Affiliation(s)
- Massimo Torreggiani
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
| | - Antioco Fois
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
| | - Claudia D’Alessandro
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.D.); (A.C.)
| | - Marco Colucci
- Unit of Nephrology and Dialysis, ICS Maugeri S.p.A. SB, Via S. Maugeri 10, 27100 Pavia, Italy;
| | | | - Adamasco Cupisti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.D.); (A.C.)
| | - Giorgina Barbara Piccoli
- Nephrology and Dialysis, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000 Le Mans, France; (A.F.); (G.B.P.)
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, 10100 Torino, Italy
| |
Collapse
|
6
|
Luteinizing Hormone Action in Human Oocyte Maturation and Quality: Signaling Pathways, Regulation, and Clinical Impact. Reprod Sci 2020; 27:1223-1252. [PMID: 32046451 PMCID: PMC7190682 DOI: 10.1007/s43032-019-00137-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022]
Abstract
The ovarian follicle luteinizing hormone (LH) signaling molecules that regulate oocyte meiotic maturation have recently been identified. The LH signal reduces preovulatory follicle cyclic nucleotide levels which releases oocytes from the first meiotic arrest. In the ovarian follicle, the LH signal reduces cyclic nucleotide levels via the CNP/NPR2 system, the EGF/EGF receptor network, and follicle/oocyte gap junctions. In the oocyte, reduced cyclic nucleotide levels activate the maturation promoting factor (MPF). The activated MPF induces chromosome segregation and completion of the first and second meiotic divisions. The purpose of this paper is to present an overview of the current understanding of human LH signaling regulation of oocyte meiotic maturation by identifying and integrating the human studies on this topic. We found 89 human studies in the literature that identified 24 LH follicle/oocyte signaling proteins. These studies show that human oocyte meiotic maturation is regulated by the same proteins that regulate animal oocyte meiotic maturation. We also found that these LH signaling pathway molecules regulate human oocyte quality and subsequent embryo quality. Remarkably, in vitro maturation (IVM) prematuration culture (PMC) protocols that manipulate the LH signaling pathway improve human oocyte quality of cultured human oocytes. This knowledge has improved clinical human IVM efficiency which may become a routine alternative ART for some infertile patients.
Collapse
|
7
|
Shamseldeen AM, Ali Eshra M, Ahmed Rashed L, Fathy Amer M, Elham Fares A, Samir Kamar S. Omega-3 attenuates high fat diet-induced kidney injury of female rats and renal programming of their offsprings. Arch Physiol Biochem 2019; 125:367-377. [PMID: 29741967 DOI: 10.1080/13813455.2018.1471511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Context: Maternal diet composition could influence fetal organogenesis. Objective: We investigated effects of high fat diet (HFD) intake alone or combined with omega 3 during pregnancy, lactation and early days of weaning on nephrogenesis of pups and maternal renal function and morphology. Material and methods: Mothers and their pups included in each group were supplied with the same diet composition. Rats were divided into group I, II and III supplied with chow of either 10 kcal%, 45 kcal% or 45 kcal% from fat together with omega-3 respectively. Results: Group II showed increased serum urea and creatinine, renal TNF-α, IL1β. Structural injury was observed in mothers and their pups as Bowman's capsule and tubular dilatation and increased expression of PCNA that were decreased following omega-3 supplementation added to down regulation of Wnt4, Pax2 gene and podocin expression. Discussion and conclusion: Omega-3 supplementation improves lipid nephrotoxicity observed in mothers and their pups.
Collapse
Affiliation(s)
| | - Mohammed Ali Eshra
- a Department of Physiology Faculty of Medicine, Cairo University , Cairo , Egypt
| | - Laila Ahmed Rashed
- b Department of Biochemistry Faculty of Medicine, Cairo University , Cairo , Egypt
| | - Marwa Fathy Amer
- b Department of Biochemistry Faculty of Medicine, Cairo University , Cairo , Egypt
| | - Amal Elham Fares
- c Department of Medical Histology Faculty of Medicine, Cairo University , Cairo , Egypt
| | - Samaa Samir Kamar
- c Department of Medical Histology Faculty of Medicine, Cairo University , Cairo , Egypt
| |
Collapse
|
8
|
Tain YL, Chan JYH, Lee CT, Hsu CN. Maternal Melatonin Therapy Attenuates Methyl-Donor Diet-Induced Programmed Hypertension in Male Adult Rat Offspring. Nutrients 2018; 10:nu10101407. [PMID: 30279341 PMCID: PMC6213858 DOI: 10.3390/nu10101407] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/15/2018] [Accepted: 09/28/2018] [Indexed: 01/03/2023] Open
Abstract
Although pregnant women are advised to consume methyl-donor food, some reports suggest an adverse outcome. We investigated whether maternal melatonin therapy can prevent hypertension induced by a high methyl-donor diet. Female Sprague-Dawley rats received either a normal diet, a methyl-deficient diet (L-MD), or a high methyl-donor diet (H-MD) during gestation and lactation. Male offspring were assigned to four groups (n = 7⁻8/group): control, L-MD, H-MD, and H-MD rats were given melatonin (100 mg/L) with their drinking water throughout the period of pregnancy and lactation (H-MD+M). At 12 weeks of age, male offspring exposed to a L-MD or a H-MD diet developed programmed hypertension. Maternal melatonin therapy attenuated high methyl-donor diet-induced programmed hypertension. A maternal L-MD diet and H-MD diet caused respectively 938 and 806 renal transcripts to be modified in adult offspring. The protective effects of melatonin against programmed hypertension relate to reduced oxidative stress, increased urinary NO₂- level, and reduced renal expression of sodium transporters. A H-MD or L-MD diet may upset the balance of methylation status, leading to alterations of renal transcriptome and programmed hypertension. A better understanding of reprogramming effects of melatonin might aid in developing a therapeutic strategy for the prevention of hypertension in adult offspring exposed to an excessive maternal methyl-supplemented diet.
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 and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan.
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan.
| | - Chien-Te Lee
- Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, 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
|
9
|
Tain YL, Hsu CN. Developmental Origins of Chronic Kidney Disease: Should We Focus on Early Life? Int J Mol Sci 2017; 18:ijms18020381. [PMID: 28208659 PMCID: PMC5343916 DOI: 10.3390/ijms18020381] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 02/03/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is becoming a global burden, despite recent advances in management. CKD can begin in early life by so-called "developmental programming" or "developmental origins of health and disease" (DOHaD). Early-life insults cause structural and functional changes in the developing kidney, which is called renal programming. Epidemiological and experimental evidence supports the proposition that early-life adverse events lead to renal programming and make subjects vulnerable to developing CKD and its comorbidities in later life. In addition to low nephron endowment, several mechanisms have been proposed for renal programming. The DOHaD concept opens a new window to offset the programming process in early life to prevent the development of adult kidney disease, namely reprogramming. Here, we review the key themes on the developmental origins of CKD. We have particularly focused on the following areas: evidence from human studies support fetal programming of kidney disease; insight from animal models of renal programming; hypothetical mechanisms of renal programming; alterations of renal transcriptome in response to early-life insults; and the application of reprogramming interventions to prevent the programming of kidney disease.
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 and Chang Gung University College of Medicine, 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
|
10
|
Boubred F, Daniel L, Buffat C, Tsimaratos M, Oliver C, Lelièvre-Pégorier M, Simeoni U. The magnitude of nephron number reduction mediates intrauterine growth-restriction-induced long term chronic renal disease in the rat. A comparative study in two experimental models. J Transl Med 2016; 14:331. [PMID: 27899104 PMCID: PMC5129242 DOI: 10.1186/s12967-016-1086-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is a risk factor for hypertension (HT) and chronic renal disease (CRD). A reduction in the nephron number is proposed to be the underlying mechanism; however, the mechanism is debated. The aim of this study was to demonstrate that IUGR-induced HT and CRD are linked to the magnitude of nephron number reduction, independently on its cause. METHODS Systolic blood pressure (SBP), glomerular filtration rate (GFR), proteinuria, nephron number, and glomerular sclerosis were compared between IUGR offspring prenatally exposed to a maternal low-protein diet (9% casein; LPD offspring) or maternal administration of betamethasone (from E17 to E19; BET offspring) and offspring with a normal birth weight (NBW offspring). RESULTS Both prenatal interventions led to IUGR and a similar reduction in birth weight. In comparison to NBW offspring, BET offspring had a severe nephron deficit (-50% in males and -40% in females, p < 0.01), an impaired GFR (-33%, p < 0.05), and HT (SBP+ 17 mmHg, p < 0.05). Glomerular sclerosis was more than twofold higher in BET offspring than in NBW offspring (p < 0.05). Long-term SBP, GFR, and glomerular sclerosis were unchanged in LPD offspring while the nephron number was moderately reduced only in males (-28% vs. NBW offspring, p < 0.05). CONCLUSION In this study, the magnitude of nephron number reduction influences long term renal disease in IUGR offspring: a moderate nephron number is an insufficient factor. Extremely long-term follow-up of adults prenatally exposed to glucocorticoids are required.
Collapse
Affiliation(s)
- Farid Boubred
- NORT, Aix-Marseille Université, INRA, INSERM, 13005, Marseille, France. .,Department of Neonatology, Hôpital la Conception, AP-HM, 147 Boulevard Baille, 13385, Marseille Cedex, France.
| | - Laurent Daniel
- UPRES EA3281, Aix-Marseille Université, 13005, Marseille, France
| | | | - Michel Tsimaratos
- Pédiatrie Multidisciplinaire-Hôpital de la Timone, Marseille, France
| | - Charles Oliver
- NORT, Aix-Marseille Université, INRA, INSERM, 13005, Marseille, France
| | | | - Umberto Simeoni
- DOHaD Laboratory, CHUV University Hospital and UNIL, Lausanne, Switzerland
| |
Collapse
|
11
|
Tain YL, Chan JYH, Hsu CN. Maternal Fructose Intake Affects Transcriptome Changes and Programmed Hypertension in Offspring in Later Life. Nutrients 2016; 8:nu8120757. [PMID: 27897982 PMCID: PMC5188412 DOI: 10.3390/nu8120757] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/01/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022] Open
Abstract
Hypertension originates from early-life insults by so-called “developmental origins of health and disease” (DOHaD). Studies performed in the previous few decades indicate that fructose consumption is associated with an increase in hypertension rate. It is emerging field that tends to unfold the nutrient–gene interactions of maternal high-fructose (HF) intake on the offspring which links renal programming to programmed hypertension. Reprogramming interventions counteract disturbed nutrient–gene interactions induced by maternal HF intake and exert protective effects against developmentally programmed hypertension. Here, we review the key themes on the effect of maternal HF consumption on renal transcriptome changes and programmed hypertension. We have particularly focused on the following areas: metabolic effects of fructose on hypertension and kidney disease; effects of maternal HF consumption on hypertension development in adult offspring; effects of maternal HF consumption on renal transcriptome changes; and application of reprogramming interventions to prevent maternal HF consumption-induced programmed hypertension in animal models. Provision of personalized nutrition is still a faraway goal. Therefore, there is an urgent need to understand early-life nutrient–gene interactions and to develop effective reprogramming strategies for treating hypertension and other HF consumption-related diseases.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 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
|
12
|
Rideau Batista Novais A, Pham H, Van de Looij Y, Bernal M, Mairesse J, Zana-Taieb E, Colella M, Jarreau PH, Pansiot J, Dumont F, Sizonenko S, Gressens P, Charriaut-Marlangue C, Tanter M, Demene C, Vaiman D, Baud O. Transcriptomic regulations in oligodendroglial and microglial cells related to brain damage following fetal growth restriction. Glia 2016; 64:2306-2320. [PMID: 27687291 DOI: 10.1002/glia.23079] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 11/06/2022]
Abstract
Fetal growth restriction (FGR) is a major complication of human pregnancy, frequently resulting from placental vascular diseases and prenatal malnutrition, and is associated with adverse neurocognitive outcomes throughout life. However, the mechanisms linking poor fetal growth and neurocognitive impairment are unclear. Here, we aimed to correlate changes in gene expression induced by FGR in rats and abnormal cerebral white matter maturation, brain microstructure, and cortical connectivity in vivo. We investigated a model of FGR induced by low-protein-diet malnutrition between embryonic day 0 and birth using an interdisciplinary approach combining advanced brain imaging, in vivo connectivity, microarray analysis of sorted oligodendroglial and microglial cells and histology. We show that myelination and brain function are both significantly altered in our model of FGR. These alterations, detected first in the white matter on magnetic resonance imaging significantly reduced cortical connectivity as assessed by ultrafast ultrasound imaging. Fetal growth retardation was found associated with white matter dysmaturation as shown by the immunohistochemical profiles and microarrays analyses. Strikingly, transcriptomic and gene network analyses reveal not only a myelination deficit in growth-restricted pups, but also the extensive deregulation of genes controlling neuroinflammation and the cell cycle in both oligodendrocytes and microglia. Our findings shed new light on the cellular and gene regulatory mechanisms mediating brain structural and functional defects in malnutrition-induced FGR, and suggest, for the first time, a neuroinflammatory basis for the poor neurocognitive outcome observed in growth-restricted human infants. GLIA 2016;64:2306-2320.
Collapse
Affiliation(s)
- Aline Rideau Batista Novais
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Réanimation et Pédiatrie Néonatales, Groupe Hospitalier Robert Debré, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Hoa Pham
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Yohan Van de Looij
- Laboratory for Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Division of Development and Growth, Department of Child and Adolescent Medicine, Geneva University Hospital and School of Medicine, Geneva, Switzerland
| | - Miguel Bernal
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Jerome Mairesse
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Elodie Zana-Taieb
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France.,Université Paris-Descartes, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, Paris, France
| | - Marina Colella
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Pierre-Henri Jarreau
- Fondation PremUp, Paris, France.,Université Paris-Descartes, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, Paris, France
| | - Julien Pansiot
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Florent Dumont
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Paris, France
| | - Stéphane Sizonenko
- Division of Development and Growth, Department of Child and Adolescent Medicine, Geneva University Hospital and School of Medicine, Geneva, Switzerland
| | - Pierre Gressens
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Christiane Charriaut-Marlangue
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Mickael Tanter
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Charlie Demene
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Daniel Vaiman
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Paris, France
| | - Olivier Baud
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France. .,Assistance Publique - Hôpitaux de Paris, Service de Réanimation et Pédiatrie Néonatales, Groupe Hospitalier Robert Debré, Paris, France. .,Université Paris Diderot, Paris, France. .,Fondation PremUp, Paris, France.
| |
Collapse
|
13
|
Zhang L, Chen W, Dai Y, Zhu Z, Liu Q. Detection of expressional changes induced by intrauterine growth restriction in the developing rat pancreas. Exp Biol Med (Maywood) 2016; 241:1446-56. [PMID: 27190278 DOI: 10.1177/1535370216638771] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/18/2016] [Indexed: 12/15/2022] Open
Abstract
Intrauterine growth retardation (IUGR) is a disorder that can result in permanent changes in the physiology and metabolism of the newborn, which increased the risk of disease in adulthood. Evidence supports IUGR as a risk factor for the development of diabetes mellitus, which could reflect changes in pancreas developmental pathways. We sought to characterize the IUGR-induced alterations of the complex pathways of pancreas development in a rat model of IUGR. We analyzed the pancreases of Sprague Dawley rats after inducing IUGR by feeding a maternal low calorie diet from gestational day 1 until term. IUGR altered the pancreatic structure, islet areas, and islet quantities and resulted in abnormal morphological changes during pancreatic development, as determined by HE staining and light microscopy. We identified multiple differentially expressed genes in the pancreas by RT-PCR. The genes of the insulin/FoxO1/Pdx1/MafA signaling pathway were first expressed at embryonic day 14 (E14). The expressions of insulin and MafA increased as the fetus grew while the expressions of FoxO1 and Pdx1 decreased. Compared with the control rats, the expressions of FoxO1, Pdx1, and MafA were lower in the IUGR rats, whereas insulin levels showed no change. Microarray profiling, in combination with quantitative real-time PCR, uncovered a subset of microRNAs that changed in their degree of expression throughout pancreatic development. In conclusion, our data support the hypothesis that IUGR influences the development of the rat pancreas. We also identified new pathways that appear to be programmed by IUGR.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Endocrinology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Wei Chen
- Department of Endocrinology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Yuee Dai
- Department of Endocrinology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Ziyang Zhu
- Department of Endocrinology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Qianqi Liu
- Department of Endocrinology, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
14
|
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
|
15
|
Renal Transcriptome Analysis of Programmed Hypertension Induced by Maternal Nutritional Insults. Int J Mol Sci 2015; 16:17826-37. [PMID: 26247937 PMCID: PMC4581224 DOI: 10.3390/ijms160817826] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 06/12/2015] [Accepted: 07/30/2015] [Indexed: 12/25/2022] Open
Abstract
Maternal nutrition can affect development, leading to long-term effects on the health of offspring. The most common outcome is programmed hypertension. We examined whether alterations in renal transcriptome are responsible for generating programmed hypertension among four different models using next-generation RNA sequencing (NGS) technology. Pregnant Sprague-Dawley rats received 50% caloric restriction (CR), intraperitoneal injection of 45 mg/kg streptozotocin, 60% high-fructose (HF) diet, or 1% NaCl in drinking water to conduct CR, diabetes, HF, or high-salt models, respectively. All four models induced programmed hypertension in adult male offspring. We observed 16 shared genes in a two-week-old kidney among four different models. The identified differential expressed genes (DEGs) that are related to the regulation of blood pressure included Adrb3, Alb, Apoe, Calca, Kng1, Adm2, Guca2b, Hba2, Hba-a2, and Ppara. The peroxisome proliferator-activated receptor (PPAR) signaling pathway and glutathione metabolism pathway were shared by the CR, diabetes, and HF models. Conclusively, a variety of maternal nutritional insults induced the same phenotype-programmed hypertension with differential alterations of renal transcriptome in adult male offspring. The roles of DEGs identified by the NGS in this study deserve further clarification to develop ideal maternal dietary interventions and thus spare the next generations from the burden of hypertension.
Collapse
|
16
|
Maternal fructose-intake-induced renal programming in adult male offspring. J Nutr Biochem 2015; 26:642-50. [DOI: 10.1016/j.jnutbio.2014.12.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/05/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022]
|
17
|
Sheen JM, Yu HR, Tiao MM, Chen CC, Huang LT, Chang HY, Tain YL. Prenatal dexamethasone-induced programmed hypertension and renal programming. Life Sci 2015; 132:41-8. [PMID: 25921765 DOI: 10.1016/j.lfs.2015.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/08/2015] [Accepted: 04/11/2015] [Indexed: 01/19/2023]
Abstract
AIMS Antenatal glucocorticoids can induce long-term effects on offspring health, including hypertension. Programmed hypertension has been observed in a prenatal dexamethasone (DEX) exposure model. However, how renal programming responds to prenatal DEX at different stages of development and the impact of DEX on programmed hypertension remain unclear. Therefore, we utilized RNA next-generation sequencing (NGS) to analyze the renal transcriptome in the offspring to examine whether key genes and pathways are responsible for DEX-induced renal programming and hypertension. MAIN METHODS Pregnant rats received intraperitoneal dexamethasone from gestational day 16 to 22. Prenatal DEX-induced programmed hypertension was examined in male offspring at 16 weeks of age. KEY FINDINGS Prenatal DEX modified 431 renal transcripts from the nephrogenesis stage to adulthood in a constant manner. At the pre-hypertensive and established hypertension stages, we identified 11 and 13 differentially expressed genes related to blood pressure regulation, respectively. Among these genes, Npr3, Ptgs2, Agt, Edn3, Ephx2, Agtr1b, and Gucy1a3 are associated with endothelium-derived hyperpolarizing and contractile factors (EDHF and EDCF). Genes in the arachidonic acid metabolism pathway may potentially be key genes contributing to programmed hypertension. In addition, DEX induced soluble epoxide hydrolase expression (Ephx2 gene encoding protein). SIGNIFICANCE Prenatal DEX elicits an imbalance between EDHFs and EDCFs that might lead to renal programming and hypertension. The arachidonic acid metabolism pathway is a common pathway contributing to programmed hypertension. Our results highlight candidate genes and pathways involved in renal programming as targets for therapeutic approaches to prevent programmed hypertension in children exposed to antenatal corticosteroids.
Collapse
Affiliation(s)
- Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Chih-Cheng Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan; Department of Traditional Chinese Medicine, Chang Gung University, Linkow, Taiwan
| | - Hsin-Yu Chang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
18
|
Zana-Taieb E, Pham H, Franco-Montoya ML, Jacques S, Letourneur F, Baud O, Jarreau PH, Vaiman D. Impaired alveolarization and intra-uterine growth restriction in rats: a postnatal genome-wide analysis. J Pathol 2015; 235:420-30. [DOI: 10.1002/path.4470] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/17/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023]
Affiliation(s)
- E Zana-Taieb
- Université Paris Descartes; Paris France
- Fondation PremUp, 53 avenue de l'Observatoire, 75014 Paris; France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1141; Paris France
- Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, 53 Avenue de l'Observatoire, 75014 Paris; France
| | - H Pham
- Fondation PremUp, 53 avenue de l'Observatoire, 75014 Paris; France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1141; Paris France
| | - ML Franco-Montoya
- Institut National de la Santé et de la Recherche Médicale (INSERM) U955 IMRB Equipe 04, Faculté de Médecine de Créteil, 94010 Créteil; France
| | - S Jacques
- Genom'ic, INSERM U1016, CNRS UMR8104, Paris; France
| | - F Letourneur
- Genom'ic, INSERM U1016, CNRS UMR8104, Paris; France
| | - O Baud
- Fondation PremUp, 53 avenue de l'Observatoire, 75014 Paris; France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1141; Paris France
- Assistance Publique - Hôpitaux de Paris, Service de Réanimation et Pédiatrie Néonatales, Hôpital Robert Debré, Paris; France
- Université Paris Diderot; Paris France
| | - PH Jarreau
- Université Paris Descartes; Paris France
- Fondation PremUp, 53 avenue de l'Observatoire, 75014 Paris; France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1141; Paris France
- Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, 53 Avenue de l'Observatoire, 75014 Paris; France
| | - D Vaiman
- Institut Cochin, INSERM U1016-CNRS, UMRS 104; Paris France
| |
Collapse
|
19
|
Zana-Taieb E, Butruille L, Franco-Montoya ML, Lopez E, Vernier F, Grandvuillemin I, Evain-Brion D, Deruelle P, Baud O, Delacourt C, Jarreau PH. Effect of two models of intrauterine growth restriction on alveolarization in rat lungs: morphometric and gene expression analysis. PLoS One 2013; 8:e78326. [PMID: 24278109 PMCID: PMC3836790 DOI: 10.1371/journal.pone.0078326] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/11/2013] [Indexed: 01/21/2023] Open
Abstract
Intrauterine growth restriction (IUGR) in preterm infants increases the risk of bronchopulmonary dysplasia, characterized by arrested alveolarization. We evaluated the impact of two different rat models (nitric oxide synthase inhibition or protein deprivation) of IUGR on alveolarization, before, during, and at the end of this postnatal process. We studied IUGR rat pups of dams fed either a low protein (LPD) or a normal diet throughout gestation and pups of dams treated by continuous infusion of Nω-nitro-L-arginine methyl ester (L-NAME) or its diluent on the last four days of gestation. Morphometric parameters, alveolar surface (Svap), mean linear intercept (MLI) and radial alveolar count (RAC) and transcriptomic analysis were determined with special focus on genes involved in alveolarization. IUGR pups regained normal weight at day 21 in the two treated groups. In the LPD group, Svap, MLI and RAC were not different from those of controls at day 4, but were significantly decreased at day 21, indicating alveolarization arrest. In the L-NAME group, Svap and RAC were significantly decreased and MLI was increased at day 4 with complete correction at day 21. In the L-NAME model, several factors involved in alveolarization, VEGF, VEGF-R1 and –R2, MMP14, MMP16, FGFR3 and 4, FGF18 and 7, were significantly decreased at day 4 and/or day 10, while the various factors studied were not modified in the LPD group. These results demonstrate that only maternal protein deprivation leads to sustained impairment of alveolarization in rat pups, whereas L-NAME impairs lung development before alveolarization. Known growth factors involved in lung development do not seem to be involved in LPD-induced alveolarization disorders, raising the question of a possible programming of altered alveolarization.
Collapse
Affiliation(s)
- Elodie Zana-Taieb
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U767, Paris, France
- PremUp, Paris, France
- Service de Médecine et Réanimation néonatales de Port-Royal, Groupe hospitalier Cochin, Broca, Hôtel-Dieu, Assistance Publique – Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Paris, France
- * E-mail:
| | - Laura Butruille
- Unité environnement périnatal et croissance, EA4489, Faculté de Médecine, Pôle recherche, IFR 114,Université Lille Nord de France, Lille, France
| | | | - Emmanuel Lopez
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U767, Paris, France
- PremUp, Paris, France
- Service de Médecine et Réanimation néonatales de Port-Royal, Groupe hospitalier Cochin, Broca, Hôtel-Dieu, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Flore Vernier
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U767, Paris, France
- PremUp, Paris, France
| | - Isabelle Grandvuillemin
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1076, Faculté de Pharmacie, Université de la Méditerranée. Marseille, France
| | - Danièle Evain-Brion
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U767, Paris, France
- PremUp, Paris, France
- Université Paris Descartes, Paris, France
| | - Philippe Deruelle
- Unité environnement périnatal et croissance, EA4489, Faculté de Médecine, Pôle recherche, IFR 114,Université Lille Nord de France, Lille, France
| | - Olivier Baud
- PremUp, Paris, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U676, Paris, France
- Service de Réanimation et Pédiatrie néonatales, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
- Université Paris Diderot, Paris, France
| | - Christophe Delacourt
- PremUp, Paris, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U955 IMRB Equipe 04, Créteil, France
- Service de Pneumologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique – Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Paris, France
| | - Pierre-Henri Jarreau
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U767, Paris, France
- PremUp, Paris, France
- Service de Médecine et Réanimation néonatales de Port-Royal, Groupe hospitalier Cochin, Broca, Hôtel-Dieu, Assistance Publique – Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Paris, France
| |
Collapse
|
20
|
Developmental origins of chronic renal disease: an integrative hypothesis. Int J Nephrol 2013; 2013:346067. [PMID: 24073334 PMCID: PMC3773449 DOI: 10.1155/2013/346067] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/17/2013] [Accepted: 07/03/2013] [Indexed: 01/06/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of mortality. Hypertension (HT) is one of the principal risk factors associated with death. Chronic kidney disease (CKD), which is probably underestimated, increases the risk and the severity of adverse cardiovascular events. It is now recognized that low birth weight is a risk factor for these diseases, and this relationship is amplified by a rapid catch-up growth or overfeeding during infancy or childhood. The pathophysiological and molecular mechanisms involved in the “early programming” of CKD are multiple and partially understood. It has been proposed that the developmental programming of arterial hypertension and chronic kidney disease is related to a reduced nephron endowment. However, this mechanism is still discussed. This review discusses the complex relationship between birth weight and nephron endowment and how early growth and nutrition influence long term HT and CKD. We hypothesize that fetal environment reduces moderately the nephron number which appears insufficient by itself to induce long term diseases. Reduced nephron number constitutes a “factor of vulnerability” when additional factors, in particular a rapid postnatal growth or overfeeding, promote the early onset of diseases through a complex combination of various pathophysiological pathways.
Collapse
|
21
|
Figueroa H, Lozano M, Suazo C, Eixarch E, Illanes SE, Carreño JE, Villanueva S, Hernández-Andrade E, Gratacós E, Irarrazabal CE. Intrauterine growth restriction modifies the normal gene expression in kidney from rabbit fetuses. Early Hum Dev 2012; 88:899-904. [PMID: 22944138 DOI: 10.1016/j.earlhumdev.2012.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/30/2012] [Accepted: 07/08/2012] [Indexed: 01/17/2023]
Abstract
The aim of this work was to study the effect of intrauterine growth restriction (IUGR) on fetal kidneys. The IUGR was induced by uteroplacental vessels ligature in a model of pregnant rabbit. We centralized the study in the gene expression of essential proteins for fetal kidney development and kidney protection against hypoxia, osmotic stress, and kidney injury. The gene expression of HIF-1α, NFAT5, IL-1β, NGAL, and ATM were studied by qRT-PCR and Western blot in kidneys from control and IUGR fetuses. Experimental IUGR fetuses were significantly smaller than the control animals (39 vs. 48 g, p<0.05). The number of glomeruli was decreased in IUGR kidneys, without morphological alterations. IUGR increased the gene expression of HIF-1α, NFAT5, IL-1β, NGAL, and ATM (p<0.05) in kidneys of fetuses undergoing IUGR, suggesting that fetal blood flow restriction produce alterations in gene expression in fetal kidneys.
Collapse
Affiliation(s)
- Horacio Figueroa
- Department of Obstetrics & Gynecology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cassidy-Bushrow AE, Wegienka G, Barone CJ, Valentini RP, Yee J, Havstad S, Johnson CC. Race-specific relationship of birth weight and renal function among healthy young children. Pediatr Nephrol 2012; 27:1317-23. [PMID: 22399075 PMCID: PMC3692279 DOI: 10.1007/s00467-012-2136-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 01/19/2012] [Accepted: 02/13/2012] [Indexed: 01/14/2023]
Abstract
BACKGROUND Low birth weight is associated with diminished renal function. However, despite African Americans being at increased risk of low birth weight and chronic kidney disease, little is known about the association between birth weight and renal function in diverse groups. We examined racial differences in the relationship of birth weight and renal function among healthy young children. METHODS Birth weight and serum creatinine data were available on 152 children (61.8% African American; 47.4% female) from a birth cohort. Estimated glomerular filtration rate (eGFR) was calculated using the bedside Schwartz equation and gender- and gestational-age-adjusted birth weight Z-scores using the US population as a reference. Race-specific linear regression models were fit to estimate the association between birth weight Z-score and eGFR. RESULTS Mean age was 1.5 ± 1.3 years at first eGFR measurement. African Americans had lower eGFR than non-African Americans (median eGFR = 82 vs. 95 ml/min per 1.73 m(2); p = 0.06). Birth weight was significantly and positively associated with eGFR among African American (p = 0.012) but not non-African American children (p = 0.33). CONCLUSIONS We provide, for the first time, evidence suggesting that birth weight is associated with renal function in African American children. Future work is needed to determine if prenatal programming helps explain racial disparities in adult health.
Collapse
Affiliation(s)
| | - Ganesa Wegienka
- Department of Public Health Sciences, Henry Ford Hospital, One Ford Place, Detroit, Michigan
| | - Charles J. Barone
- Department of Pediatrics, Henry Ford Hospital, One Ford Place, Detroit, Michigan
| | - Rudolph P. Valentini
- The Carman and Ann Adams Department of Pediatrics, Division of Nephrology and Hypertension, Children’s Hospital of Michigan, Wayne State University School of Medicine, Detroit, Michigan
| | - Jerry Yee
- Division of Nephrology and Hypertension, Department of Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, Michigan
| | - Suzanne Havstad
- Department of Public Health Sciences, Henry Ford Hospital, One Ford Place, Detroit, Michigan
| | - Christine Cole Johnson
- Department of Public Health Sciences, Henry Ford Hospital, One Ford Place, Detroit, Michigan
| |
Collapse
|
23
|
Vaiman D, Gascoin-Lachambre G, Boubred F, Mondon F, Feuerstein JM, Ligi I, Grandvuillemin I, Barbaux S, Ghigo E, Achard V, Simeoni U, Buffat C. The intensity of IUGR-induced transcriptome deregulations is inversely correlated with the onset of organ function in a rat model. PLoS One 2011; 6:e21222. [PMID: 21731679 PMCID: PMC3120850 DOI: 10.1371/journal.pone.0021222] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/23/2011] [Indexed: 01/21/2023] Open
Abstract
A low-protein diet applied during pregnancy in the rat results in intrauterine growth restricted (IUGR) fetuses. In humans, IUGR is associated with increased perinatal morbidity, higher incidence of neuro-developmental defects and increased risk of adult metabolic anomalies, such as diabetes and cardiovascular disease. Development and function of many organs are affected by environmental conditions such as those inducing fetal and early postnatal growth restriction. This phenomenon, termed "fetal programming" has been studied unconnectedly in some organs, but very few studies (if any) have investigated at the same time several organs, on a more comparative basis. However, it is quite probable that IUGR affects differentially most organ systems, with possible persistent changes in gene expression. In this study we address transcriptional alterations induced by IUGR in a multi-organ perspective, by systematic analysis of 20-days rat fetuses. We show that (1) expressional alterations are apparently stronger in organs functioning late in foetal or postnatal life than in organs that are functioning early (2) hierarchical classification of the deregulations put together kidney and placenta in one cluster, liver, lungs and heart in another; (3) the epigenetic machinery is set up especially in the placenta, while its alterations are rather mild in other organs; (4) the genes appear deregulated in chromosome clusters; (5) the altered expression cascades varies from organ to organ, with noticeably a very significant modification of the complement and coagulation cascades in the kidney; (6) we found a significant increase in TF binding site for HNF4 proteins specifically for liver genes that are down-regulated in IUGR, suggesting that this decrease is achieved through the action of HNF transcription factors, that are themselves transcriptionnally induced in the liver by IUGR (x 1.84 fold). Altogether, our study suggests that a combination of tissue-specific mechanisms contributes to bring about tissue-driven modifications of gene cascades. The question of these cascades being activated to adapt the organ to harsh environmental condition, or as an endpoint consequence is still raised.
Collapse
Affiliation(s)
- Daniel Vaiman
- Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Cochin, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Simeoni U, Ligi I, Buffat C, Boubred F. Adverse consequences of accelerated neonatal growth: cardiovascular and renal issues. Pediatr Nephrol 2011; 26:493-508. [PMID: 20938692 DOI: 10.1007/s00467-010-1648-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 07/12/2010] [Accepted: 07/26/2010] [Indexed: 12/13/2022]
Abstract
Epidemiological and experimental studies show that the risk of cardiovascular and metabolic diseases at adulthood is inversely related to the weight at birth. Although with less evidence, low birth weight has been suggested to increase the risk of chronic kidney disease (CKD). It is well established that the developmental programming of arterial hypertension and of renal disease involves in particular renal factors, especially nephron endowment, which is reduced in low birth weight and maternal diabetes situations. Experimental studies, especially in rodents, have demonstrated the long-term influence of postnatal nutrition and/or postnatal growth on cardiovascular, metabolic and renal functions, while human data are scarce on this issue. Vascular and renal diseases appear to have a "multihits" origin, with reduced nephron number the initial hit and rapid postnatal growth the second hit. This review addresses the current understanding of the role of the kidney, both as a mechanism and as a target, in the developmental origins of adult disease theory, with a particular focus on the long-term effects of postnatal growth and nutrition.
Collapse
Affiliation(s)
- Umberto Simeoni
- Division of Neonatology, Hôpital la Conception, Assistance Publique-Hôpitaux de Marseille, 147 Boulevard Baille, 13385, Marseille, France.
| | | | | | | |
Collapse
|
25
|
Lapillonne A, Razafimahefa H, Rigourd V, Granier M. La nutrition du prématuré. Arch Pediatr 2011; 18:313-23. [DOI: 10.1016/j.arcped.2010.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 11/10/2010] [Accepted: 12/14/2010] [Indexed: 10/18/2022]
|
26
|
Hepatic expression of the GH/JAK/STAT/IGF pathway, acute-phase response signalling and complement system are affected in mouse offspring by prenatal and early postnatal exposure to maternal high-protein diet. Eur J Nutr 2011; 50:611-23. [DOI: 10.1007/s00394-011-0168-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 01/10/2011] [Indexed: 12/20/2022]
|
27
|
Ritz E, Amann K, Koleganova N, Benz K. Prenatal programming-effects on blood pressure and renal function. Nat Rev Nephrol 2011; 7:137-44. [PMID: 21283139 DOI: 10.1038/nrneph.2011.1] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Impaired intrauterine nephrogenesis-most clearly illustrated by low nephron number-is frequently associated with low birthweight and has been recognized as a powerful risk factor for renal disease; it increases the risks of low glomerular filtration rate, of more rapid progression of primary kidney disease, and of increased incidence of chronic kidney disease or end-stage renal disease. Another important consequence of impaired nephrogenesis is hypertension, which further amplifies the risk of onset and progression of kidney disease. Hypertension is associated with low nephron numbers in white individuals, but the association is not universal and is not seen in individuals of African origin. The derangement of intrauterine kidney development is an example of a more general principle that illustrates the paradigm of plasticity during development-that is, that transcription of the genetic code is modified by epigenetic factors (as has increasingly been documented). This Review outlines the concept of prenatal programming and, in particular, describes its role in kidney disease and hypertension.
Collapse
Affiliation(s)
- Eberhard Ritz
- Division of Nephrology, Department of Internal Medicine, University of Heidelberg, Heidelberg 69100, Germany.
| | | | | | | |
Collapse
|
28
|
Simeoni U, Boubred F, Buffat C, Grandvuillemin I, Ligi I. [Risk for long term disease in low birth weight infants]. Arch Pediatr 2010; 17:669-70. [PMID: 20654834 DOI: 10.1016/s0929-693x(10)70052-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- U Simeoni
- Service de Néonatologie, Pôle Parents-Enfants, Hôpital de La Conception, Assistance Publique, Hôpitaux de Marseille et UMR 608 INSERM & Fondation Santé, Sport et Développement Durable, Université de la Méditerranée, Marseille, France.
| | | | | | | | | |
Collapse
|
29
|
Auer J, Camoin L, Guillonneau F, Rigourd V, Chelbi ST, Leduc M, Laparre J, Mignot TM, Vaiman D. Serum profile in preeclampsia and intra-uterine growth restriction revealed by iTRAQ technology. J Proteomics 2010; 73:1004-17. [DOI: 10.1016/j.jprot.2009.12.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/14/2009] [Accepted: 12/30/2009] [Indexed: 12/24/2022]
|
30
|
Fauque P, Mondon F, Letourneur F, Ripoche MA, Journot L, Barbaux S, Dandolo L, Patrat C, Wolf JP, Jouannet P, Jammes H, Vaiman D. In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model. PLoS One 2010; 5:e9218. [PMID: 20169163 PMCID: PMC2821408 DOI: 10.1371/journal.pone.0009218] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 01/24/2010] [Indexed: 12/17/2022] Open
Abstract
Background Assisted Reproductive Technologies (ART) are increasingly used in humans; however, their impact is now questioned. At blastocyst stage, the trophectoderm is directly in contact with an artificial medium environment, which can impact placental development. This study was designed to carry out an in-depth analysis of the placental transcriptome after ART in mice. Methodology/Principal Findings Blastocysts were transferred either (1) after in vivo fertilization and development (control group) or (2) after in vitro fertilization and embryo culture. Placentas were then analyzed at E10.5. Six percent of transcripts were altered at the two-fold threshold in placentas of manipulated embryos, 2/3 of transcripts being down-regulated. Strikingly, the X-chromosome harbors 11% of altered genes, 2/3 being induced. Imprinted genes were modified similarly to the X. Promoter composition analysis indicates that FOXA transcription factors may be involved in the transcriptional deregulations. Conclusions For the first time, our study shows that in vitro fertilization associated with embryo culture strongly modify the placental expression profile, long after embryo manipulations, meaning that the stress of artificial environment is memorized after implantation. Expression of X and imprinted genes is also greatly modulated probably to adapt to adverse conditions. Our results highlight the importance of studying human placentas from ART.
Collapse
Affiliation(s)
- Patricia Fauque
- Service d'Histologie-Embryologie, Biologie de la Reproduction, Hôpital Cochin, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fauque P, Ripoche MA, Tost J, Journot L, Gabory A, Busato F, Le Digarcher A, Mondon F, Gut I, Jouannet P, Vaiman D, Dandolo L, Jammes H. Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos. Hum Mol Genet 2010; 19:1779-90. [DOI: 10.1093/hmg/ddq059] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Boubred F, Daniel L, Buffat C, Feuerstein JM, Tsimaratos M, Oliver C, Dignat-George F, Lelièvre-Pégorier M, Simeoni U. Early postnatal overfeeding induces early chronic renal dysfunction in adult male rats. Am J Physiol Renal Physiol 2009; 297:F943-51. [PMID: 19656908 DOI: 10.1152/ajprenal.90704.2008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Low birth weight is associated with an increased risk of hypertension and renal dysfunction at adulthood. Such an association has been shown to involve a reduction of nephron endowment and to be enhanced by accelerated postnatal growth in humans. However, while low-birth-weight infants often undergo catch-up growth, little is known about the long-term vascular and renal effects of accelerated postnatal growth. We surimposed early postnatal overfeeding (OF; reduction of litter size during the suckling period) to appropriate-birth-weight (NBW+OF) and intrauterine growth restriction (IUGR; IUGR+OF) pups, obtained after a maternal gestational low-protein diet. Blood pressure (systolic blood pressure; SBP) and renal function (glomerular filtration rate; GFR) were measured in young and aging offspring. Glomerulosclerosis and nephron number were determined in aging offspring (22 mo). Nephron number was reduced in both IUGR and IUGR+OF male offspring (by 24 and 26%). GFR was reduced by 40% in 12-mo-old IUGR+OF male offspring, and both NBW+OF and IUGR+OF aging male offspring had sustained hypertension (+25 mmHg) and glomerulosclerosis, while SBP and renal function were unaffected in IUGR aging offspring. Female offspring were unaffected. In conclusion, in this experimental model, early postnatal OF in the neonatal period has major long-lasting effects. Such effects are gender dependent. Reduced nephron number alone, associated with IUGR, may not be sufficient to induce long-lasting physiological alterations, and early postnatal OF acts as a "second hit." Early postnatal OF is a suitable model with which to study the long-term effects of postnatal growth in the pathogenesis of vascular disorders and renal disease.
Collapse
Affiliation(s)
- Farid Boubred
- INSERM UMR608, Faculté de Pharmacie, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Diabetes in pregnancy has been shown to induce long-term effects in offspring. While considerable attention is focused on the increased incidence of type 2 diabetes mellitus (T2DM) in adult offspring from diabetic mothers, cardiovascular alterations, including hypertension, are also part of lifelong consequences of in-utero exposure to increased glucose concentrations. This review examines the epidemiologic and mechanistic issues involved in the developmental programming of long-term consequences in offspring of diabetic mothers, with a particular emphasis on the renal and vascular mechanisms of hypertension. The factors of increased incidence of T2DM and of obesity in adults born after exposure to diabetes during pregnancy are also discussed, as evidence is accumulating that a vicious circle involving lifelong consequences of diabetes in pregnancy in offspring contributes to the current worldwide epidemic of T2DM.
Collapse
Affiliation(s)
- Umberto Simeoni
- INSERM UMR608, Université de la Méditerranée, France; Faculté de Médecine, Université de la Méditerranée, France; Division of Neonatology, Assistance Publique-Hôpitaux de Marseille, France.
| | | |
Collapse
|
34
|
Abdel-Hakeem AK, Henry TQ, Magee TR, Desai M, Ross MG, Mansano RZ, Torday JS, Nast CC. Mechanisms of impaired nephrogenesis with fetal growth restriction: altered renal transcription and growth factor expression. Am J Obstet Gynecol 2008; 199:252.e1-7. [PMID: 18639218 PMCID: PMC2932650 DOI: 10.1016/j.ajog.2008.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 03/01/2008] [Accepted: 05/24/2008] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Maternal food restriction during pregnancy results in growth-restricted newborns and reduced glomerular number, contributing to programmed offspring hypertension. We investigated whether reduced nephrogenesis may be programmed by dysregulation of factors controlling ureteric bud branching and mesenchyme to epithelial transformation. STUDY DESIGN At 10 to 20 days' gestation, Sprague Dawley pregnant rats (n = 6/group) received ad libitum food; food-restricted rats were 50% food restricted. At embryonic day 20, messenger ribonucleic acid (mRNA) and protein expression of Wilms' tumor 1 gene product (WT1), paired box transcription factor (Pax)-2, fibroblast growth factor (FGF)-2, glial cell line-derived neurotrophic factor (GDNF), cRET, wingless-type mouse mammary tumor virus integration site (WNT)4, WNT11, bone morphogenetic protein (BMP)-4, BMP7, and FGF7 were determined by real-time polymerase chain reaction and Western blotting. RESULTS Maternal food restriction resulted in up-regulated mRNA expression for WT1, FGF2, and BMP7, whereas Pax2, GDNF, FGF7, BMP4, WNT4, and WNT11 mRNAs were down-regulated. Protein expression was concordant for WT1, GDNF, Pax2, FGF7, BMP4, and WNT4. CONCLUSION Maternal food restriction altered gene expression of fetal renal transcription and growth factors and likely contributes to development of offspring hypertension.
Collapse
Affiliation(s)
- Ahmed K Abdel-Hakeem
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|